CA2862289C - Modulators of methyl modifying enzymes, compositions and uses thereof - Google Patents

Modulators of methyl modifying enzymes, compositions and uses thereof Download PDF

Info

Publication number
CA2862289C
CA2862289C CA2862289A CA2862289A CA2862289C CA 2862289 C CA2862289 C CA 2862289C CA 2862289 A CA2862289 A CA 2862289A CA 2862289 A CA2862289 A CA 2862289A CA 2862289 C CA2862289 C CA 2862289C
Authority
CA
Canada
Prior art keywords
methyl
piperidin
alkylene
compound
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2862289A
Other languages
French (fr)
Other versions
CA2862289A1 (en
Inventor
Brian K. Albrecht
James Edmund Audia
Andrew S. Cook
Les A. Dakin
Martin Duplessis
Victor S. Gehling
Jean-Christophe Harmange
Christopher G. Nasveschuk
Rishi G. Vaswani
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Constellation Pharmaceuticals Inc
Original Assignee
Constellation Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Constellation Pharmaceuticals Inc filed Critical Constellation Pharmaceuticals Inc
Publication of CA2862289A1 publication Critical patent/CA2862289A1/en
Application granted granted Critical
Publication of CA2862289C publication Critical patent/CA2862289C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Agents for modulating methyl modifying enzymes, compositions and uses thereof are provided herein. Such agents may be represented by compounds having structural formula (see formula I) or a pharmaceutically acceptable salts thereof. Such compounds may be used in the treatment of cancer.

Description

MODULATORS OF METHYL MODIFYING ENZYMES, COMPOSITIONS AND USES
THEREOF
[0001]
BACKGROUND OF THE INVEr=FTION
Eukaryotic chromatin is composed of macromolecular complexes called nucleosomes. A nucleosome has 147 base pairs of DNA wrapped around a protein octatner having two subunits of each of histone protein H2A, H2B, I-13, and H4. Histone proteins are subject to post-translational modifications which in turn affect chromatin structure and gene expression. One type of post-translational modification found on histones is methylation of lysine and arginine residues. Histone methylation plays a critical role in the regulation of gene expression in eukaryotes. Methylation affects chromatin structure and has been linked to both activation and repression of transcription (Zhang and Reinberg, Genes Dev.
15:2343-2360, 2001). Enzymes that catalyze attachment and removal of methyl groups from histones are implicated in gene silencing, embryonic development, cell proliferation, and other processes.
SUMMARY OF THE INVEN'llON
[0002] The present disclosure encompasses the recognition that methyl modifying enzymes are an attractive target for modulation, given their role in the regulation of diverse biological processes. It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as agents that stimulate activity of histone methyl modifying enzymes, including histone methylases and histone demethyiases. Such compounds have the general formula I:

Rx n\
Z N
Ri 0 or a pharmaceutically acceptable salt thereof, wherein each variable is as defined herein.
100031 Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with a methyl modifying enzyme. Such diseases, disorders, or conditions include those described herein.
100041 Compounds provided by this invention are also useful for the study of methyl modifying enzymes in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by methyl modifying enzymes and the comparative evaluation of new methyl modifying enzyme modulators.
BRIEF DESCRIPTION OF THE DRAWINGS
100051 Figure 1. Exemplary compounds of formulae 1 and IL
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
I. General Description of Compounds (lithe Invention [00061 In certain embodiments, the present invention provides a compound of formula I:

RLRX
(Th or a pharmaceutically acceptable salt thereof, wherein:
Z is =C(R2)- or =N-;
each of XI and X2 is independently selected from =N-, and =C(R3)-;

X3 is selected from =N-, and =C(R6)-;
no more than one of X', X2, and X3 is =N-;
each R1 and R2 is independently selected from hydrogen, halo, -OH, -CN, CI-CI
alkyl, -0-(C1-C4 alkyl), -N(R)2, -(Co-C4 alkylene)-aryl, -(Co-C4 alkylene)-heteroaryl, -(Co-C4 alkylene)-heterocyclyl, and -(Co-C4 a1kylene)-carbocycly1; or one R1 and R2 are taken together with atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocyclyl ring;
each R3 and R6 is independently selected from hydrogen, halo, -CN, -(Co-C4 alkylene)-R8, C6 alkenyl or alkyny1)-R9, -(C1-C4 alkylene)-0-R9, -(C1-C4 alkylene)-0-(C1-C4 a1kylene)-R8, -0-(Co-C4 a1kylene)-R9, -0-(C2-C4 alkylene)-0-R8, -0-(C1-C4 al kylene)-R9, -(Co-C4 alkylene)-N(R7)2, -(Co-C4 alkylene)-C(0)-0-1e, -(Co-a1kylene)-0-C(0)-R.9, -(Co-C4 alk.ylene)-C(0)-N(R7)2, -(Co-C4 alkylene)-N(R9)-C(0)-le, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 alkylene)-N(R9)-C(0)-(R7), -(Co-C4 alky1ene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(Co-C4 alky1erte)-S(0)2-N(R7)2; or two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocycly1 fused to the ring comprising X', X2 and X3;
It' is Q, -S(0)2-Q, -C(0)---Q, or Q is selected from aryl, heteroaryl, .heterocyclyl, or carbocyclyl;
R4 is selected from C2-C6 alkyl, -CH2-0-(C1-C4 alkyl) and -(Co-C6 alkylene)-Q, wherein one or two methylene units in the alkyl or alkylene portion of R4 are optionally and independently replaced by -0-, -S -WO) -, -S(:=0)2-, or -N(111 )-; or one methylene unit of R4 is taken together with X2 or X3, when the X2 or X3 is =C(R3)-, and the intervening atoms to form a heteroaryl or heterocyclyl fused to the ring comprising X', X', and X3;
R5 is selected from hydrogen, -(Co-C6 allcylene)-Q, and C1-C6 alkyl, wherein one or two methylene units in R5 are optionally and independently replaced by -0-, -S-, -S(=0) -> -S(=0)2-, or -Nle-;
each R' is independently selected from -(Co-C4 alkylene)-R9, -(Co-C4 alkylene)-0-R9, -S(0)2-R8, -C(=0)-R8, -C(=0)-N(R9)2, -(C1-C4 a1kylene)-0-C(=0)-and -(Co-C4 alkylene)-C(=0)-0-R9; or
3 two le are taken together with the nitrogen atom to which they arc conunonly bound to form an optionally substituted heterocycly1 or heteroaryl;
R8 is selected from C1-C4 alkyl, aryl, heteroaryl, carbocyclyl and heterocycly1;
R9 is selected from hydrogen or R8:
R1 is selected from hydrogen, CI-C4 alkyl, -S(=0)2-R9, -C(=0)-R8, -C(=0)-N(R9)(R12), and -C(=0)-0-R";
R11 is selected from unsubstituted C 1- C4 alkyl and C1-C4 haloalky I;
R12 is selected from hydrogen, unsubstituted C1-C4 alkyl and C,-C4 haloalkyl;
and wherein unless otherwise designated any alkyl, alkylene, alkcnyl, allcynyl, aryl, hetcroaryl, heterocyclyl or carbocyclyl portion of the compound is optionally substituted.
2. Compounds and Definitions 100071 Definitions of specific functional groups and chemical terms arc described in more detail below. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed., inside cover, and specific functional groups are generally defined as described therein.
Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Organic Chemisiiy, Thomas Sorrell, University Science Books, Sausalito, 1999; Smith and March March's Advanced Organic Chemistry, .5(1' Edition, John Wiley & Sons, Inc., New York, 2001; Larock, Comprehensive Organic Transformations, VC1-1 Publishers, Inc., New York, 1989; Carruthers, Some Modern Method of Organic Synthesis, 3rd Edition, Cambridge University Press, Cambridge, 1987.
[00081 Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomerie, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastercomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only
4 in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.
Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention.
[00091 Where a particular enantiomer is preferred, it may, in some embodiments be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched." "Optically-enriched," as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer. In certain embodiments the compound is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer.
Preferred enantiomers may be isolated from. racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPI,C) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses. See, for example, Jacques et al., Enantiomers, Racemates and Resolutions (Wiley Intemience, New York, 1981); Wilen, et al., Tetrahedron 33:2725 (1977); Eliel, E.L.
Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN
1972).
[00101 A wavy bond ) at a chiral center in a chemical structure is used to denote compounds of the invention that are optically pure, but whose optical rotation has not been determined. A straight bond at a chiral center indicates a racemic mixture although, as stated above, the invention also includes all possible isomeric forms of the racemate.
[00111 The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quatemized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-211-pyrroly1), NH (as in pyffolidinyl) or NW (as in N-substituted pyrrolidinyl)).
100121 As used herein a "direct bond" or "covalent bond" refers to a single, double or triple bond. In certain embodiments, a "direct bond" or "covalent bond" refers to a single bond.
[00131 The terms "halo" and "halogen" as used herein refer to an atom selected from fluorine (fluoro,-F), chlorine (chloro,-CI), bromine (bromo,-Br), and iodine (iodo,-I).

[00141 The term "aliphatic" or "aliphatic group", as used herein, denotes a hydrocarbon moiety that may be straight-chain (i.e., unbranched), branched, or cyclic (including fused, bridging, and spiro-fused polycyclic) and may be completely saturated or may contain one or more units of unsaturation, but which is not aromatic. Unless otherwise specified, aliphatic groups contain 1-6 carbon atoms. In some embodiments, aliphatic groups contain 1-4 carbon atoms, and in yet other embodiments aliphatic groups contain 1-3 carbon atoms.
Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof such as (cycloalkyl)alkyl, (cycloallcenyl)alkyl or (cycloalkyl)alkenyl.
[00151 The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[00161 As used herein, the term "bivalent C1_8 (or C14 saturated or unsaturated, straight or branched, hydrocarbon chain", refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[00171 The term "alkylene" refers to a bivalent alkyl group. An "alkylene chain" is a polymethylene group, i.e.,-(CH2).-, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[00181 The term "alkenylene" refers to a bivalent alkenyl group. A
substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
100191 The term "alkynylene" refers to a bivalent alkynyl group.
100201 The term "methylene unit" refers to a divalent -CH2- group present in an alkyl, alkenyl, alkynyl, alkylene, alkenylene, or allcynylene moiety.
[00211 The term "Co alkylene" as used herein means a bond. Thus, a moiety defined herein as "-(Co-C6 alkylene)-aryl" includes both -aryl (i.e., Co allcylene-aryl) and -(C1-C6 alkylene)-aryl.
100221 The term "alkyl," as used herein, refers to a monovalent saturated, straight- or branched-chain hydrocarbon radical derived from an aliphatic moiety containing between one and six carbon atoms by removal of a single hydrogen atom. In some embodiments, alkyl contains 1-5 carbon atoms. In another embodiment, alkyl contains 1-4 carbon atoms. In still other embodiments, alkyl contains 1-3 carbon atoms. In yet another embodiment, alkyl contains 1-2 carbons. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, sec-pentyl, iso-pentyl, tert-butyl, n-pentyl, neopentyl, n-hexyl, sec-hexyl, and the like.
[00231 The term "alkenyl," as used herein, denotes a monovalent group derived from a straight- or branched-chain aliphatic moiety having at least one carbon-carbon double bond by the removal of a single hydrogen atom. In certain embodiments, alkenyl contains 2-6 carbon atoms. In certain embodiments, alkenyl contains 2-5 carbon atoms. In some embodiments, alkenyl contains 2-4 carbon atoms. In another embodiment, alkenyl contains 2-3 carbon atoms.
Alkenyl groups include, for example, ethertyl ("vinyl"), propenyl ("ally1"), butenyl, 1 -methyl-2-buten- 1 -yl , and the like.
[00241 The term "alkynyl," as used herein, refers to a monovalent group derived from a straight- or branched-chain aliphatic moiety having at least one carbon-carbon triple bond by the removal of a single hydrogen atom. In certain embodiments, alkynyl contains 2-6 carbon atoms.
In certain embodiments, alkynyl contains 2-5 carbon atoms. In some embodiments, alkynyl contains 2-4 carbon atoms. In another embodiment, alkynyl contains 2-3 carbon atoms.
Representative alkynyl groups include, but are not limited to, ethynyl, 2-propyrtyl ("propargyr), 1-propynyl, and the like.
[00251 The term "carbocycly1" (also referred to herein as "carbocycle"
"cycloaliphatic" or "cycloalkyl"), as used herein, means a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but where there is no ring is aromatic.
[00261 The term "aryl" used alone or as part of a larger moiety as in "aralkyr, "arallcoxy", or "atyloxyalkyr, refers to monocyclic and bicyclic carbon ring systems having a total of five to 10 ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members. The term "aryl" may be used interchangeably with the term "aryl ring". In certain embodiments of the present invention, "aryl"
refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term "aryl", as it is used herein, is a group in which an aromatic ring is fused to one or more carbocyclyl rings regardless of whether the aromatic carbon ring or the carbocyclic ring is the pendant ring, or a group in which an aromatic carbon ring is fused to one or more heteroaryl or heterocyclyl, rings, such as indanyl, phthalimidyl, naphthimidyl, phenantriidinyl, or tetrahydronaphthyl, and the like, wherein the pendant ring of the fused ring system is the aromatic carbon ring.
[00271 The terms "heteroaryl" and "heteroar-", used alone or as part of a larger moiety, e.g., "heteroaralkyl", or "heteroaralkoxy", refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 It electrons shared in a cyclic array;
and having, in addition to carbon atoms, from one to five heteroatoms. The term "heteroatom" refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quatemized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms "heteroaryl" and "heteroar-", as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, wherein the pendant ring of the fused ring system is heteroaromatic. Non-limiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, 5,6,7,8-tetrahydroquinolinyl, 5,6,7,8-tetrahydroisoquinolinyl, and pyrido[2,3-13]-1,4-oxazin-3(4H)-one. A heteroaryl group may be mono- or bicyclic. The term "heteroaryl."
may be used interchangeably with the terms "heteroaryl ring", "heteroaryl group", or "heteroaromatic", any of which terms include rings that are optionally substituted. The term "heteroaralkyl" refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. The term.
"heteroarylene" refers to a bivalent mono- or bicyclic heteroaryl ring.
[00281 As used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic radical", and "heterocyclic ring" are used interchangeably and refer to a stable 4- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. In certain embodiments, a "heterocycle", group is a 1,1'-heterocyclylene group (i.e., a spiro-fused ring). When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrroly1), NH (as in pyrrolidinyl), or '7,\IR (as in N-substituted pyrrolidinyl).
[0029] A
heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl, 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms "heterocycle", "heterocyclyl", "heterocyclyl ring", "heterocyclic group", "heterocyclic moiety", and "heterocyclic radical", are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, 2-azabicyclo[2.2.1]heptanyl, octahydroindolyl, or tetrahydroquinolinyl , wherein the pendant ring of the fused ring system is heterocyclyl. A heterocyclyl group may be mono- or bicyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
[0030i As used herein, the term "partially unsaturated" refers to a ring moiety that includes at least one double or triple bond between ring atoms but is not aromatic. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[00311 As used herein, the terms "carbocyclylene" or "cycloalkylene" are used interchangeably and refer to a bivalent carbocyclyl or cycloalkyl group. In certain embodiments, a carbocyclylene or cycloalkylene group is a 1,1-cycloallcylene group (i.e., a spiro-fused ring).
Exemplary 1,1-cycloalkylene groups include , 0 or . In other embodiments, a cycloalkylene group is a 1,2-cycloalkylene group or a 1,3-cycloalkylene group.

>0 411,1_ \43 =/ 34:15.2(4, Exemplary 1,2-eyeloalkylene groups include V, 0 and _________________ i .
Exemplary 11- C ) 1,3-cycloalkylene groups include and [0032] As described herein, compounds of the invention may contain "optionally substituted" moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substitucnt selected from a specified group, the substituent may be either the. same or different at each position.
Combinations of substituents envisioned under this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[0033]
Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; -(CH2)0_4Ir; -(CH2)040W; -0-(CH2)0-4C(0)0R ; -(012)0.40-1(OR )2; -(C142)0.4SR ; -(CF12)0.4Ph, which may be substituted with R ; -(CH2)a.40(CH2)0.1Ph which may be substituted with 1r; -CH=CHPh, which may be substituted with R ; -NO2; -CN; -N3; 4CH2)o-4N(Rc)2; -(CH2)G-4N(R. )C(0)1r; -N(Rc)C(S)R ; -(C112)0.4N(R
)C(0)NR*2; -N(R )C
(S)NR 2; -(C112)0-4N(R )C(0)0.1V; -N(R )N(R1C(0)R ; -N(R1N(R )C(0)NR 2; -N(R
)N(R ) C7(0)0R ; -(CH2)0-4C(0)R ; -C(S)R ; -(CF12.)o-4C(0)0R ; -(CH2)o-4C(0)SR ; -(CH2)0-4C(0)0Si R 3; -(CH2)0.40C(0)R ; -0C(0)(CH2)0.4SR-; -SC(S)SR ; -(CH2)0.4SC(0)Rc;
-(CH2)o-4C(0)NR 2; -C(S)NR 2; -C(S)SR*; -SC(S)S1r; -(CH2)0_40C(0)NR'2; -C(0)N(OR )R :
-C(0)C(0)R ; -C(0)CH2C(0)R ; -C(NOR")R ; -(CH2)0-4SSR ; -(CH2)04S(0)2R ; -(CH2)0-4S(0) 20R ; -(CH2)0.4.0S(0)2R ; -S(0)2NR. 2; -(CH2)0.4S(0)R ; -N(R )S(0)21 R 2; -N(11 )S(0)2R ; -N
(01r)1r; -C(NH)NR 2; -P(0)21r; -P(0)R 2; -0P(0)R 2; -0P(0)(0102; -SiR 3; -(C1_4 straight or branched alkylene)O-N(Ir)2; or -(C1..4 straight or branched alkylene)C(0)0-N(R)2, wherein each R may be substituted as defined below and is independently hydrogen, C1-aliphatic, -CH2Ph, -0(0-12)0.1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of 1r, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono-or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Suitable monovalent substituents on R (or the ring formed by taking two independent occurrences of ir together with their intervening atoms), are independently halogen, -(CH2)0_21e, -(halon, -(CH2)o-20H, -(CH2)0-201e, -(CH2)0_2CH(0R.)2; -0(halon, -CN, -N3, -(CH2)o-2C(0)R., -(CH2)0_2C(0)0H, -(CH2)0_2C(0)0e, -(CH2)0-2SR., -(CH2)o-2SH. -(CH2)0-2NH2, -(CH2)0_2NHR*, -(CH2)0_2NR`2, -NO2, -SiR.3, -0SiR."3, -C(0)SR., 4C14 straight or branched alkylene)C(0)01e, or -SSW wherein each R. is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from C1-4 aliphatic, -CH2Ph, -0(CH2)0_1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
Suitable divalent substituents on a saturated carbon atom of R include =0 and =S.
(00351 Suitable divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR*2, =NNHC(0)R*, =NNHC(0)01e, -NNHS(0)2R*, -NOR*,-0(C(R*2))2-30-, or-S(C(R*2))2.-3S-, wherein each independent occurrence of R* is selected from hydrogen, C1_6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include:-0(CR*2)2-30-, wherein each independent occurrence of R* is selected from hydrogen, C1_6 aliphatic which may be substituted as defmed below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00361 Suitable substituents on the aliphatic group of R* include halogen, -R., -(halon, -OH, -0(halor), -EN, -C(0)0H, -C(0)0R*, -NH2, -NH1e, -NR
.2, or -NO2, wherein each R.* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C14 aliphatic, -CH2Ph, -0(CH2)0.1Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00371 Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include-Rt,11.2, -C(0)1e, -C(0)0Rt, -C(0)C(0)Rt, -C(0)CH2C(0)Rt, -S(0)2Rt, -S(0)2NRt2, -C(S)NRt2, -C(NH)NRt2, or -N(Rt)S(0)2Rt; wherein each Rt is independently hydrogen, C1-6 aliphatic which may be substituted as defined below, unsubstituted -0Ph, or an unsubstituted
5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of Rt, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from. nitrogen, oxygen, or sulfur.
[00381 Suitable substituents on the aliphatic group of Rt are independently halogen, 42, -(haloR.*), -OH, -0R.=, -0(haloR*), -CN, -C(0)0H, -C(0)0R*, -NH2, -NHR.', -NR
.2, or -NO2, wherein each R. is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci..4a1iphatic, -CH2Ph, -0(CH2)0.11Ph, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[00391 As used herein, the term "inhibitor" is defined as a compound that binds to and /or inhibits a target S-adenosylmethionine (SAM) utilizing enzyme with measurable affinity. In certain embodiments, an inhibitor has an IC50 and/or binding constant of less about 50 1AM, less than about 11.1M, less than about 500 nM, less than about 100 nM, or less than about 10 nM.
[00401 The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in activity of at least one SAM utilizing enzyme between a sample comprising a provided compound, or composition thereof, and at least one SAM
dependent enzyme, and an equivalent sample comprising at least one SAM dependent enzyme, in the absence of said compound, or composition thereof.
3. Description of Exemplary Compounds [00411 In certain embodiments, the present invention provides a compound of formula I:

LON R"

Ri 0 or a pharmaceutically acceptable salt thereof, wherein each variable is as defined above and described herein. This same structure may also be represented as:
R" N 0 xl--X2 Z.L,\N R x R a 0 to distinguish between the two R' moieties attached to the left-hand ring.
100421 As defined generally above and herein, Z is =C(R2)- or =N-, wherein R2 is as defined above and described herein. In some embodiments, Z is =C(R2) - wherein R2 is as defined above and described herein. In some embodiments, Z is =CH-. In some embodiments, Z
is =N-.
[00431 As defined generally above and herein, each of X1 and X2 is independently selected from =N- and =C(R3)-, wherein R3 is as defined above and described herein. In some embodiments, each of X1 and X2 is independently =C(R3)-, wherein R3 is as defmed above and described herein. In some embodiments, X1 is =C(R3)- and X2 is =N-, wherein R3 is as defined above and described herein. In some embodiments, X1 is selected from =CH-, =C(CI-C4 alkyl)- and =C(ary1)-, and X2 is =N-. In some embodiments, X' is selected from =CH-, =C(CH3)- and =C(pheny1)- and X2 is =N-, wherein the phenyl is optionally substituted. In som.e embodiments, X1 is =C(H)- and X2 is =N-. In some embodiments, X.1 is =C(CH3)-and X2 is =N.
In some embodiments, X2 is =C(R3)- and X.1 is =N-, wherein R3 is as defined above and described herein. In some embodiments, X2 is =C(H)- and X1 is =N-. In some embodiments, X2 is =C(CH3)- and X1 is =N-. In some embodiments, X1 is selected from =CH-, =C(CI-C4 alkyl)- and =C(ary1)-. In some embodiments, X2 is =N-.
[00441 In some embodiments, each of X1 and X2 is independently =C(R3)-, wherein two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, heterocycl.yl, or carbocyclyl fused to the ring comprising X1, X2 and X.
wherein each of X1, X2 and X3 is as defined above and described herein. In some embodiments, each of X and X2 is independently =C(R3)-, wherein two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, or carbocyclyl fused to the ring comprising Xi, X2 and X3, wherein each of Xl, X2 and X3 is as defined above and described. herein.
[0045] As defined generally above and herein, X3 is independently selected from =N- and =C(R6)-, wherein R6 is as defined above and described herein.
[0046] In some embodiments, X3 is =N-.
[0047] In some embodiments, X3 is =C(R6)-, wherein R6 is as defined above and described herein. In some embodiments, X3 is selected from =C(CI-C4 alkyl)-, =CH-, =C(OH)-, =C(CN)-, =C(0-C1-C4 alkyl), =C(C(0)-N(102)-, =C(aryI)-, =C(carbocycly1)-, and =C(heterocycly1)-, wherein each R7 is independently as defined above and described herein. In some embodiments, X3 is selected from =CH-, =C(CH3)-, =C(CH2CH3)-, =C(OH)-, =C((N)-, =C(OCH3)-, =C(C(0)NH2)-, =C(cyclopropy1)-, (phenyl.)-, and =C(oxetany1)-, wherein the cyclopropyl, phenyl or oxetanyl is optionally substituted. In some embodiments, X3 is selected from =CH-, =C(CH3)-, =C(CH2CH3)-, =C(OH)-, =C(CN)-, =C(0-CH3), =C(C(0)-NH2)-, =C(cyclopropy1)-, =C(phenyI)-, and =C(oxetany1)-, wherein the cyclopropyl, phenyl or oxetanyl is substituted. In some embodiments, X3 is selected from =CH-, =C(013)-, :=C((H2C113)-, =C(OFI)-, :::C(CN)_, =C(0-CH3), =C(C(0)-NEI2)-, =C(cyclopropy1)-, =C(phenyI)-, and =C(oxetany1)-, wherein the cyclopropyl, phenyl or oxetanyl is unsubstituted. In some embodiments, X3 is selected from =CH-, =C(CH3)-, or =C(phenyI)-, wherein the phenyl is optionally substituted.
100481 As defined generally above and herein, each It' and R2 is independently selected from hydrogen, halo, -OH, -CN, alkyl, -0-(C1-C4 alkyl), -N(R7)2, -(Co-C4 alkylene)-aryl, -(Co-C4 alkylene)-heteroaryl, -(Co-C4 alkylene)-heterocyclyl, and -(Co-C4 alkylene)-carbocyclyl; or one R1 and R2 are taken together with atoms to which they are bound to form an atyl, heteroaryl, heterocyclyl, or carbocyclyl ring;
wherein each R7 is independently as defined above and described herein.
100491 In some embodiments, each R1 and R2 is hydrogen. In one embodiment, each R1 and R2 is independently selected from halo, -OH, -CN, C1-C4 alkyl, -0-(C1-C4 alley!), -N(R7)2, C4 allcylene)-aryl, -(C0-C4 alkylene)-heteroaryl, -(Co-C4 alleylene)-heterocyclyl, and -(C0-C4 alkylene)-carbocyclyl; or one R' and R2 are taken together with atoms to which they are bound to form an aryl, heterowyl, heterocyclyl, or carbocyclyl ring;
wherein each R7 is independently as defined above and described herein.
[00501 In some embodiments, each RI and R.2 is independently selected from halo, -OH, -CN, C1-C4 alkyl, -0-(C1-C4 alkyl), -N(R.7)2, -(Co-C4 alkylene)-aryl, -(Co-C4 alkylene)-heteroaryl, -(Co-C4 allcylene)-heterocyclyl, and -(C0-C4 alkylene)-carbocyclyl, wherein each R7 is independently as defined above and described herein. In some embodiments, one R1 and R2 are taken together with atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocyclyl ring.
[0051.1 In some embodiments, each R.' is independently selected from hydrogen and -CH3.
In some embodiments, each R' is hydrogen. In some embodiments, each R' is -CH3. In som.e embodiments, one is hydrogen. In some embodiments, one R' is -CH3. In some embodiments, one RI is -CH3 and the other RI is selected from -0-CH3 and -NH-[00521 In some embodiments, each of RI and R2 is hydrogen. In som.e embodiments, each RI is -CH3; and Z is =C(H)-. In some embodiments, one RI is -CH3; the other RI
is -0-CH3 or -NH-CH3 ; and Z is =C(H)-.
100531 In some embodiments, R2 is hydrogen.
[00541 In some embodiments, one R.' is -CFII and the other RI is selected from -C1-C2 alkyl and -0-(C1-C2 alkyl), wherein RI is optionally substituted with one or more fluor . In one aspect of this embodiment R.11) is -C113. In another aspect of this embodiment RI' is selected from -0CH3, -C113, -OCHF2, and -CH2CH3. In a more specific aspect of this embodiment Rib is -CH3 and Z is =CH-. In an even more specific aspect of this embodiment is selected from -OCH3, -CH3, -OCHF2, and -CH2CH3; Rib is -CH3; and Z is =CH-.
[00551 As defined generally above and herein, each R3 and R6 is independently selected from hydrogen, halo, -CN, -(Co-C4 alkylene)-R8, -(C2-C6 allcenyl or alkyny1)-R9, -(C1-C4 allcylene)-0-R9, -(C1-C4 alkylene)-0-(Cl-C4 allcylene)-R8, -0-(Co-C4 alkylene)-R9, -0-(C2-C4 alkylene)-0-R8, -0-(C1-C4 allcylene)-R9, -(Co-C4 allcylene)-N(R7)2, -(Co-C4 allcylenc)-C(0)-0-R9, -(Co-C4 alkylene)-0-C(0)-R9, -(Co-C4 alkylene)-C(0)-N(R7)2, -(Co-C4 alkylene)-N(R9)-C(0)-R9, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 allcylene)-N(R9)-C(0)-(R7), -(Co-C4 alkylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(C0-C4 alkylene)-S(0)2-N(R7)2; or two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocyclyl fused to the ring comprising XI, X2 and X3;
wherein each of XI, X2, X3, R7, R8 and R9 is independently as defined above and described herein.
[00561 In some embodiments, each R3 and R6 is independently selected from hydrogen, halo, -EN, -(Co-C4 alkylene)-R8, -(C2-C6 alkenyl or alkynyI)-R9, -(C1-C4 alkylene)-0-R8, -(C1-C4 alkylene)-0-(CI-C4 alkylcne)-R8, -0-(Co-C4 alkylenc)-R9, -0-(C2-C4 alkylenc)-0-R8, -0-(C1-C4 alkylene)-R9, -(Co-C4 alkylene)-N(R7)2, -(Co-C4 alkylene)-C(0)-0-R9, -(Co-C4 alkylene)-0-C(0)-R9, -(Co-C4 alkylene)-C(0)-N(R7)2, -(Co-C4 alkylene)-N(R9)-C(0)-R9, -0-(CI-C4 alkyl ene)-C(0)-N(R7)2, -0-(C2-C4 alkylene)-N(R9)-C(0)-(R), -(Co-C4 alkylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(Co-C4 alkylene)-S(0)2-N(R9)2; or two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, heterocycl.yl, or carbocyclyl fused to the ring comprising XI, X2 and X3;
wherein each of XI, X2, X3, R7, R8 and R9 is independently as defined above and described herein.
[0057i In some embodiments, each R3 and R6 is independently selected from hydrogen, halo, -CN, -(C0-C4 alkylenc)-R8, -(C2-C6 alkenyl or alkynyl.)-R9, -(C1-C4 alkylenc)-0-R9, -(C1-C4 alkylene)-0-(C1-C4 alkylene)-R8, -0-(Co-C4 alkylene)-R9, -0-(C2-C4 alkylene)-0-R.8, -0-(C1-C4 alkylene)-R9, -(Co-C4 alkylene)-N(R7)2, -(C0-C4 alkylene)-E(0)-0-R9, -(C0-C4 alkylene)-0-C(0)-R9, -(Co-C4 alkylene)-C(0)-N(R7)2, -(Co-C4 alkylene)-N(R9)-C(0)-R9, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 alky I ene)-N(R9)-C(0)-(R7), -(C0-C4 alkylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(Co-C4 alkylene)-S(0)2-N(R7)2, wherein each of R7, R8 and R9 is independently as defined above and described herein.
[00581 In some embodiments, each R3 is independently hydrogen.
100591 In some embodiments, one R3 is hydrogen and the other R3 is independently selected from halo, -CN, -(C0-C4 alkylene)-R8, -(C2-C6 alkenyl or alkynyI)-R9, -(C1-C4 alkylene)-0-R9, -(C1-C4 alkylene)-0-(Ci-C4 alkylene)-R8, -0-(Co-C4 alkylene)-R9, -0-(C2-C4 alkylenc)-0-R8, -0-(C1-C4 alkylenc)-R9, -(Co-C4 alkylenc)-N (R7)2, -(Co-C4 alkylene)-C(0)-0-R9, -(C0-C4 alkylene)-0-C(0)-R9, -(C0-C4 alkylene)-C(0)-N(R7)2, -(C0-C4 alkylene)-N(R9)-C(0)-R9, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 alkylene)-N(R9)-C(0)-(R7), -(Co-C4 alkylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(Co-C4 alkylene)-S(0)2-N(102, wherein each of le, R8 and R9 is independently as defined above and described herein.
[00601 In some embodiments, one R3 is -(C0-C4 alkylene)-R8, wherein R8 is as defined above and described herein. In some embodiments, one R3 is R8, wherein R8 is as defined above and described herein. In some embodiments, one R3 is aryl. In some embodiments, one R3 is optionally substituted phenyl. In some embodiments, one R3 is unsubstituted phenyl. In some embodiments, one R3 is substituted phenyl. In some embodiments, one R3 is CJ-C4 alkyl. In some embodiments, one R3 is methyl. In some embodiments, R3 is 3-methoxy.
[00611 In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocyclyl fused to the ring comprising X', X2 and X3, wherein each of X', X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, or carbocyclyl fused to the ring comprising XI, X2 and X3, wherein each of Xl, X2 and X3 is independently as defined above and described herein.
[00621 in some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an aryl fused to the ring comprising XI, X2 and X3, wherein each of Xl, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted phenyl fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an unsubstituted phenyl fused to the ring comprising X', X2 and X3, wherein each of X', X? and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form a substituted phenyl fused to the ring comprising Xl, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein.
In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an aryl fused to the ring comprising XI, X2 and X3; wherein X3 is selected from =CH-, =C(C113)-, or =C(pheny1)-, wherein the phenyl is optionally substituted; and wherein each of XI, and X2 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an aryl fused to the Ri3 RÃ
ring comprising XI, X2 and X3, wherein the fused ring has the structure:
wherein R6 is as defined herein; and RI3 is selected from hydrogen, halo, phenyl, pyridinyl, and -0-(C1-C4 alkyl).
[00631 In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form a heteroaryl fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 arc taken together with the carbon atoms to which they are bound to form an optionally substituted pyrazinyl, pyrimidinyl or pyridyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In som.e embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an unsubstituted pyrazinyl, pyrimidinyl or pyridyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form. a substituted pyrazinyl, pyrimidinyl or pyridyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted pyridyl ring fused to the ring comprising X.I, X2 and X3, wherein each of XI, X2 and X.3 is independently as defined above and described herein.
In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted pyridazinyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein.
In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted pyrimidinyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein.
In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted pyrazinyl ring fused to the ring comprising XI, X2 and X3, wherein each of XI, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form a pyridyl fused to the ring comprising X1, X2 and X3, wherein the fused ring has the structure:

wherein R6 is as defined herein; and R13 is selected from hydrogen, halo, phenyl, pyridinyl, and -0-(C1-C4 alkyl).
[00641 In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form, a heterocyclyl fused to the ring comprising X1, X2 and X3, wherein each of X1, X2 and X3 is independently as defined above and described herein.
100651 In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form a carbocyclyl fused to the ring comprising X1. X2 and X3, wherein each of X1, X2 and X3 is independently as defined above and described herein. In some embodiments, two R3 are taken together with the carbon atoms to which they are bound to form a cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl ring fused to the ring comprising X1, X2 and X3, wherein each of X1, X2 and X3 is independently as defined above and described herein.
In some embodiments, two R3 are taken together with. the carbon atoms to which they are bound to form a cyclopentyl ring fused to the ring comprising X1, X2 and X3, wherein each of X1, X2 and X3 is independently as defined above and described herein.
[00661 In some embodiments, R6 is selected from hydrogen, halo, -CN, -(Co-C4 alkylene)-R.8, -(C2-C6 alkenyl or alkyilyl)-R9, -(C1-C4 a1kylene)-O-R9, -(C1-alkylene)-0-(CI-C4 allcylene)-R8, -0-(Co-C4 allcylene)-R9, -0-(C2-C4 alkylene)-0-R8, -0-(C1-C4 alkylene)-le, -(Co-C4 alkylene)-N(R7)2, -(C0-C4 alkylene)-C(0)-0-R9, -(Co-C4 alkylene)-0-C(0)-R9, -(Co-C4 alkylene)-C(0)-N(R7)2, -((o-C4 allcylene)-N(R9)-C(0)-R9, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 alkylene)-N(R9)-C(0)-(R7), -(C0-C4 alkylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(Co-C4 alkylene)-S(0)2-N(R7)2, wherein each of R7, R8 and R9 is independently as defined above and described herein.
100671 In some embodiments, R6 is hydrogen. In some embodiments, R6 is selected from halo, -CN, -(C0-C4 allcylene)-R8, -(C2-C6 al.kenyl or alkynyI)-R9, -(C1-C4 alkylene)-0-R9, -(C1-C4 alkylene)-0-(CI-C4 allcylene)-R8, -0-(Co-C4 alkylene)-R9, -0-(C2-C4 alkylene)-0-R8, -0-(C1-C4 alkylene)-W, -(Co-C4 alkylene)-N(R7)2, -(Co-C4 alkylene)-C(0)-0-R9, -(Co-C4 alkylene)-0-C(0)-R9, -(Co-C4 allcylene)-C(0)-N(R7)2, -(Co-C4 alkylene)-N(R9)-C(0)-R9, -0-(C1-C4 alkylene)-C(0)-N(R7)2, -0-(C2-C4 alky1ene)-N(R9)-C(0)-(R7), -(Co-C4 allcylene)-S(0)-R8, -(Co-C4 alkylene)-S(0)2-R8 and -(C0-C4 alkylene)-S(0)2-N(117)2, wherein each of le, R8 and R9 is independently as defined above and described herein.
[0068] In some embodiments, R6 is -O-(C2-C4 alkylene)-N(R9)-C(0)-(R7), wherein each of R7 and R9 is independently as defined above and described herein. In some embodiments, R6 is -0-(C2-C4 alkylene)-N(R9)-C(0)-(Co_C4 alkylene)-R9, wherein each R9 is independently as defined above and described herein.
[0069] In some embodiments, R6 is selected from hydrogen and -(Co-C4 alkylene)-R, wherein R.8 is as defined above and described herein. In some embodiments, R6 is selected from hydrogen and R.8, wherein R8 is as defined above and described herein. In some embodiments, R6 is selected from hydrogen, methyl and optionally substituted phenyl.
100701 In some embodiments, R6 is -(Co-C4 alkylene)-R8, wherein R8 is as defined above and described herein. In some embodiments, R6 is R6, wherein R8 is as defined above and described herein.
100711 In some embodiments. R6 is C1-C4 alkyl. In some embodiments, R6 is methyl. In some embodiments, R6 is ethyl.
[0072] In some embodiments, R6 is aryl. In some embodiments, R6 is optionally substituted phenyl. In some embodiments, R6 is substituted phenyl. In some embodiments, R6 is unsubstituted phenyl.
100731 In some embodiments, R6 is heteroaryl.
100741 In some embodiments, R6 is heterocyclyl. In some embodiments, R6 is tetrahydro-211-pyranyl, tetrahydrofuranyl, or oxetanyl. In some embodiments, R6 is oxetanyl. In some embodiments, R6 is 3-oxetanyl. In some embodiments, R6 is piperidinyl, pyrrolidinyl, azetidinyl, or aziridinyl.
100751 In some embodiments, R6 is carbocyclyl. In some embodiments, R6 is cyclopropyl.
In some embodiments, R6 is cyclobutyl. In some embodiments, R6 is cyclopentyl.
In some embodiments, R6 is cyclohexyl.

100761 In some embodiments, R6 is halo, in some embodiments, R6 is -CN. In some embodiments, R6 is -(Co-C4 alkylene)-C(0)-N(R7)2, wherein each R7 is independently as defined above and described herein. In some embodiments, R6 is -CONH2.
[00771 In some embodiments, R6 is -0-(C0-C4 alkylene)-R9, wherein R9 is as defined above and described herein. In some embodiments, R6 is -OH. In some embodiments, R6 is -OCH3.
[00781 In some embodiments, R6 is selected from C1-C4 alkyl, hydrogen, -OH, -CN, -04 C1-C4 alkyl), -C(0)N(R7)2, aryl, carbocyclyl and hcterocyclyl, wherein each le is independently as defined above and described herein. In some embodiments, R6 is hydrogen, -CH3, -CH2CH3, -OH, -CN, -OCH3, -C(0)NH2, cyclopropyl, phenyl or oxetanyl, wherein the cyclopropyl, phenyl or oxetanyl is optionally substituted. In some embodiments, R6 is hydrogen, -CH3, -CH2CH3, -OH, -CN, -OCH3, -C(0)NH2, cyclopropyl, phenyl or oxetanyl, wherein the cyclopropyl, phenyl or oxetanyl is substituted. In some embodiments, R6 is hydrogen, -CH3, -CH2CH3, -OH, -CN, -OCH3, -C(0)NH2, cyclopropyl, phenyl or oxetanyl, wherein the cyclopropyl, phenyl or oxetanyl is unsubstituted.
xl¨x2 r-\
[00791 Exemplary )(3 are depicted below.
.N -N.I,s ......,N, p--- 1%1-1- N -1- N-1.
9 ,=

/ .--1,-----...-/
01 ....,N
'NI- A, 0 0 ('N
`N.. r ......f)rs, N-1- l pil-""N

:
r"--) ,'"----(...,2\ ir-.
, , s A i N
ND, .µ-... , .N--/- NI' *'.1µ1N-1- N-1-0- CN CONH2 b '0 N=''''...=
ft N
N¨P,-1 Cs..õ--1,\==

- -= x : N-1- .4 µ..._ r \1.... -c,-A, ---o 1, NA- it i --"J\
ii-.) A,9=z.- "\---A
\
wherein R13 is selected from hydrogen, halo, phenyl, pyridinyl, and -0-(C1-C4 alkyl).
[0080] As defined generally above and herein, le is Q. -S(0)2-Q, -C(0)¨Q, or -CH(R4)(R5), wherein each of Q, R4 and R5 is independently as defined above and described herein.
[0081] In some embodiments, le is Q or -CH(R4)(R5).
100821 In some embodiments, Rx is Q wherein Q is as defined above and described herein.
In some embodiments, le is aryl. In some embodiments, le is optionally substituted phenyl. In some embodiments, le is =substituted phenyl. In some embodiments, le is substituted phenyl.
In some embodiments, le is phenyl substituted with branched or straight chain Ci-C6 alkyl. In some embodiments, le is phenyl substituted with methyl. In some embodiments, le is 2-methylphenyl. In some embodiments, le is heteroaryl. In some embodiments, R1 is pyridyl. In some embodiments, le is 2-pyridinyl. In some embodiments, le is 3-pyridinyl.
In som.e embodiments, le is 4-pyridinyl. In some embodiments, fe is carbocyclyl. In some embodiments, le is heterocyclyl. In some embodiments, le is optionally substituted tetrahydropyranyl. In some embodiments, it" is substituted tetrahydropyranyl.
In some embodiments, le is unsubstituted tetrahydropyranyl. In some embodiments, fe is aryl, heterocyclyl or heteroaryl.
[0083] In some embodiments, R" is -CH(R4)(R5), wherein each of R4 and R5 is independently as defined above and described herein.
[0084] In some embodiments, le is -S(0)2-Q, wherein Q is defined above and described herein. in some embodiments, le is-S(0)2-phenyl.
100851 In some embodiments, le is -C(0)-Q, wherein Q is defined above and described herein. in some embodiments, le is-C(0)-phenyl.

[0086] As defined generally above and herein, Q is selected from aryl, heteroaryl, heterocyclyl and carbocyclyl. In some embodiments, Q is aryl. In some embodiments, Q is heteroaryl. In some embodiments, Q is heterocyclyl. In some embodiments, Q is carbocyclyl.
[00871 As defined generally above and herein, R4 is selected from C2-C6 alkyl, -CH2-0-(C1-C4 alkyl) and -(Co-C6 alkylene)-Q, wherein one or two methylene units in the alkyl or alkylene portion of R4 are optionally and independently replaced by -0-, -S -S(=0) or -N(RI )-; or one methylene unit of R4 is taken together with X2 or X3, when the X2 or X3 is =C(R3)-, and the intervening atoms to form a heteroaryl or heterocyclyl fused to the ring comprising Xl, X2, and X3;
wherein each of Q, X', X2, X3, R3 and Rl is independently as defined above and described herein.
[00881 In some embodiments, R4 is selected from C2-C6 alkyl, -CH2-0-(C1-C4 alkyl) and -(Co-C6 alkylene)-Q, wherein one or two methylene units in the alkyl or alkylene portion of R4 are optionally and independently replaced by -0-, -S -S(=0) -S(=0)2-, or -N(111 )-; and wherein each of Q and RI is independently as defined above and described herein. In some embodiments, R4 is C2-C6 alkyl. In some embodiments, R4 is ethyl. In some embodiments, R4 is -(Co-C6 alkylene)-Q, wherein Q is as defined above and described herein. In some embodiments, R4 is Q, wherein Q is as defined above and described herein. In some embodiments, R4 is aryl. In some embodiments, R4 is optionally substituted phenyl. In some embodiments, R4 is unsubstituted phenyl. In some embodiments, R4 is substituted phenyl. In some embodiments, R4 is -(Co-C6 alkylene)-Q wherein Q is as defined above and described herein. In some embodiments, R4 is benzyl. In some embodiments, R4 is -(Co-C2 alkylene)-Q
wherein Q is as defined above and described herein. In some embodiments, R4 is -CH2-phenyl, wherein the phenyl is optionally substituted. in some embodiments, R4 is -CH2-phenyl, wherein the phenyl is substituted. In some embodiments, R4 is -CFI2-phenyl, wherein the phenyl is unsubstituted. In some embodiments. R4 is -(C0-C2 alkylene)-aryl. In some embodiments, R4 is -(Co-C2 allcylene)-heterocyclyl. In some embodiments, R4 is -(Co-C2 alkylene)-heteroaryl. In some embodiments, R4 is -(Co-C2 alkylene)-carbocyclyl.
[00891 In some embodiments, R4 is selected from C2-C6 alkyl and -(Co-C6 alkylene)-Q, wherein Q is as defined above and described herein. In some embodiments, R4 is selected from C2-C6 alkyl, -(Co-C2 alkylene)-aryl, -(Co-C2 alkylene)-heterocyclyl, and -(Co-alkylene)-heteroaryl.
[0090] In some embodiments, one methylene unit of R4 is taken together with X2 or X3, when the X2 or X3 is =C(R3)-, and the intervening atoms to form a heteroaryl or heterocyclyl fused to the ring comprising X', X2, and X3; wherein each of Q, XI, X2, X3, R3 and RI is independently as defined above and described herein.
[0091] In some embodiments, one methylene unit in the alkyl or alkylene portion of R4 is optionally replaced by -N(RI )-, wherein RI is as defined above and described herein.
[0092] In some embodiments, the alkyl or alkylene portion of R4 is optionally substituted with =0. In some embodiments, one methylene unit in the alkyl or alkylene portion of R4 is replaced by -N(R1 )- and the methylene unit next to the -N(R I )- is substituted with =0 to form -C(0)N(R10)-, wherein R11) is as defined above and described herein.
[0093] In some embodiments R4 is selected from -(C1-C3 alkylene)-0-(Ci-C2 alkyl), 1-substituted-pipieridin-4-yl, C3-C6 cycloallc.y1 optionally substituted with one or more fluor , and tetrahydropyranyl. In one aspect of this embodiment, R4 is selected from -CH2OCH3, -CH(CH3)0CH3, 4,4-d i fluorocyclohexyl , cycl opropyl tetraybyTd opyran-4-yl, I -(t-butoxycarbony1)-piperidin-4-yl, 1 -(i sobutox ycarbony1)-piperidin-4-y1 , 1 -(isopropoxycarbony1)-piperidin-4-yl, I -(2-fluoroethyl)-piperidin-4-yl, 1 -(2,2-difluoroethyl)-piperidin-4-yl, 1-(2,2,2-trifluoroethyl)-piperidin-4-yl, 1-(2-hydroxyisobuty1)-piperidin-4-yl, 1 -(hydroxyisopropylcarbony1)-piperidin-4-yl, 1 -(ethoxycarbonylmethyp-piperidin-4-yl, 1-(isopropy1 carbony1)-piperidi n-4-y1 , 1 -methylpiperidin-4-yl, 1 -(methylsulfonyI)-piperidin-4-yl, -(ethylsulfony1)-piperidin-4-yl, 1-(isopropylsulfony1)-piperidin-4-yl, 1-(pheny1)-piperidin-4-yl, 1-(oxetan-3-yl)piperidin-4-yl, 1-(pyridin-2-yI)-piperidin-4-yl, and 1-(pyrimidin-2-y1)-piperidin-4-Yl=
[0094] As defined generally above and herein, R5 is selected from hydrogen, -(Co-C6 alkylene)-Q, and C1-C6 alkyl, wherein one or two methylene units in R5 are optionally and independently replaced by -0-, -S-, -S(=0) -S(=0)2-, or -N11.1 -, wherein each o f Q and RI is independently as defined above and described herein.
[0095] In some embodiments, R5 is hydrogen. In some embodiments, R5 is selected from -(Co-C6 alkylene)-Q and C)-C6 alkyl, wherein one or two methylene units in R5 are optionally and independently replaced by -0-, -S-, -S(=0) -S(=0)2-, or -Ne-, wherein each of Q and Ri is independently as defined above and described herein. In some embodiments, R5 is C1-C6 alkyl. In some embodiments, R5 is methyl. in some embodiments, R5 is ethyl. In some embodiments, R5 is aryl. In some embodiments, R5 is optionally substituted phenyl. In some embodiments, R5 is unsubstituted phenyl. In some embodiments, R5 is substituted phenyl. In some embodiments. R5 is -(Co-C6 alkylene)-Q wherein Q is as defined above and described herein. In some embodiments, R5 is benzyl.
100961 In some embodiments, one methylene unit in the alkyl or alkylene portion of le is optionally replaced by -N(RI )-, wherein RI is as defined above and described herein.
[00971 In some embodiments, the alkyl or alkylene portion of R5 is optionally substituted with =0. In some embodiments, one methylene unit in the alkyl or alkylene portion of R5 is replaced by -N(R1 )- and the methylene unit next to the -N(R.I )- is substituted with =0 to form -C(0)N(R10)-, wherein R' is as defined above and described herein.
[00981 In some embodiments, IV is aryl, beterocycly1 or heteroaryl. In some embodiments, Rx is -CH(R4)(R5), wherein R4 is selected from C2-C6 alkyl, -(Co-C2 alkylene)-aryl, -(Co-C2 alkylene)-heterocyclyl and -(Co-C2 alkylene)-heteroaryl; and wherein R5 is selected from hydrogen and methyl.
100991 In some embodiments, IV is optionally substituted phenyl, or tetrahydropyranyl. In some embodiments, R.' is -CH(114)(R5), wherein R4 is selected from. -0-12CH3, -phenyl, and -CH2-phenyl.; and wherein R5 is selected from hydrogen and methyl.
1001001 Exemplary Rx are depicted below.

NI
õA
6 :
lb (Is =
-190101j As defined generally above and herein, each le is independently selected from -(Co-C4 allcylene)-R9, -(Co-C4 alkylene)-0-R9, -S(0)2-R8; -C(=0)-R8, -C(=0)-N(R9)2, -(C1-C4 alkylene)-0-C(-0)-R8 and -(Co-C4 alkylene)-C(=0)-0-R9; or two R7 are taken together with the nitrogen atom to which they are commonly bound to form an optionally substituted heterocyclyl or heteroaryl ring;
wherein each of R8 and R9 is independently as defined above and described herein.
[001021 In some embodiments, each R7 is independently selected from -(Co-C4 alkylene)-R9, -(Co-C4 alkylene)-0-R9, -S(0)2-R8, -C(=0)-R8, -C(=0)-N(R9)2, alkylene)-0-C(=0)-R8 and -(Co-C4 a1kylene)-C(=0)-0-R9, wherein each of R8 and R9 is independently as defined above and described herein. In some embodiments, two R7 are taken together with the nitrogen atom. to which they are commonly bound to form an optionally substituted heterocyclyl or heteroaryl. In some embodiments, each le is independently -(Co..C4 alkylene)-R9, wherein R9 is as defined above and described herein. In som.e embodiments, each R7 is independently R9, wherein R9 is as defined above and described herein.
[001031 As defined generally above and herein, R8 is selected from CI-CA
alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl. In some embodiments, R8 is CI-CI
alkyl. In some embodiments, R8 is methyl. In some embodiments, R8 is aryl. In some embodiments, R8 is optionally substituted phenyl. In some embodiments, R8 is unsubstituted phenyl. In some embodiments, R8 is substituted phenyl. In some embodiments, R8 is heteroaryl.
In som.e embodiments, R8 is carbocyclyl. In some embodiments, R8 is heterocyclyl.
[001041 As defined generally above and herein, R9 is selected from hydrogen and R.8, wherein R8 is as defined above and described herein. In some embodiments, R9 is hydrogen. In some embodiments, R9 is R8, wherein R8 is as defined above and described herein. In some embodiments, R9 is C1-C4 alkyl. In some embodiments, R9 is aryl. In some embodiments, R9 is heteroaryl. In some embodiments, R9 is carbocyclyl. In some embodiments, R9 is heterocyclyl.
[001051 As defined generally above and herein, R1 is selected from hydrogen, alkyl, -S(=0)2-R9, -C(=0)-R8, -C(=0)-N(R9)(R12), and -C(=0)-0-R11, wherein each of R8, R9, R" and R12 is independently as defined above and described herein. In some embodiments, R1 is selected from hydrogen. In some embodiments, R1 is CI-CI alkyl. In some embodiments, Rm is -S(=0)2-R9, wherein R9 is as defined above and described herein. In some embodiments, R1 is -C(=0)-R8 wherein R8 is as defined above and described herein. In some embodiments, Rm is -C(=0)-N(R9)(R12) wherein each of R9 and R12 is independently as defined above and described herein. In some embodiments, R1 is -C(=0)-0-R11 wherein R11 is as defined above and described herein.
1001061 As defined generally above and herein, RH is selected from unsubstituted C1-C4 alkyl and C1-C4 haloalkyl. In some embodiments, RH is unsubstituted C1-C4 alkyl. In some embodiments, R11 is CI-Ca haloalkyl.
[001071 As defined generally above and herein, 1112 is selected from hydrogen, unsubstituted C1-C4 alkyl and C1-C4 haloalkyl. In some embodiments, R12 is hydrogen. In some embodiments, R12 is unsubstituted C1-C4 alkyl. In some embodiments, RI2 is C1-C4 haloalkyl.
1001081 Unless otherwise designated, any alkyl, alkylene, alkenyl, alkenylene, alkynyl, alkynylene, aryl, heteroaryl, heterocyclyl or carbocyclyl portion of the compound is optionally substituted.
[001091 It will be understood by those of skill in the art that the compounds of the invention are limited to compounds that are stable. R4 and/or R5 moieties formed by replacing two methylene units with certain combinations of -0 -S-, -S(=0) -S(=0)2-, or -NR1 -are not within the scope of the present invention if the structures formed are not stable. For example, compounds wherein the R4 and/or R5 moiety comprises an -0-, -S-, -S(0)-, -S(0)2, or adjacent to an -0-, -S-, -S(0)-, -S(0)2, or -N(RI )- are not within the scope of the present invention, except for an -S(0)2- adjacent to a -N(R1 )-. In addition, neither R4, nor R5 should comprise -0-C(11)2-0-, -N-C(R8)2-0-, or -0-C(R)2-N- if the structures formed are not stable.
1001101 Unless otherwise stated, all tautom.eric forms of the compounds of the invention are XI¨X2 r.\\
N 4....S/N-1.
within the scope of the invention. In some embodiments, X') is OH . In some xl=x2 OH

\
N
embodiments, `x3 is 0 . In some embodiments, X3 r is X3 S

X1¨x2 N 4-4)(3*N-1-In some embodiments, X3 is In some embodiments, .1^^-N
¨ X2 Xl¨ X2 / OH
N Th`
3 ,Nrr In some embodiments, X3 X is is 1 Cl s' Me 11% /

frle c= 0 NH

1001111 In certain embodiments, a compound of formula I is not H
[00112.1 In certain embodiments of a compound of formula I:
when X3 is =N-, X2 is --C(CH3)-. X1 is =C(H)-, and le is 2-fluorophenyl; then R1 and R2 are fs not taken together with atoms to which they are bound to form or 3- -when each R1 is methyl, Z is =C(H)-, each of X2 and X3 is =C(CH3)-, and X1 is =C(H)-; then R.K is other than unsubstituted cyclohexyl, benzyl, pyridin-3-yl, or pyridin-2-y1;
when each R1 is methyl, Z is =C(H)-, X3 is =N-, and le is phenyl or 4-fluorophenyl; then the R.3 of X1 and the R3 of X2 are not taken together to form. unsubstitutcd C5-C7 cycloalkyl fused to th.e ring comprising X1, X2 and X3;

2&*
when XI is R5 is hydrogen, and R.4 is taken together with X3 to form wherein "1" represents the portion of the ring bound to X2, and "2" represents the portion of the ring bound to the ring carbon adjacent to X3; then X2 is other than =C(cyclopropy1)-, =C(C(C1-13)3)-, or =C(C112CF1(013)2)-, wherein the cyclopropyl is unsubstituted;
when X2 is =N-, X3 is =C(H)-, each RI is methyl, Z is =C(H)-, and Rx is 4-methylphenyl, unsubstituted phenyl, or unsubstituted benzyl; then XI is other than =C(3-methylpheny1)-, =C(3-methoxypheny1)-, =C(phenyl)-, =C(4-chlorophenyl), =C(thien-2-y1.)-, or =C(pyridin-3-y1); and when X2 is =N-, Xi is =C(11)-, each RI is methyl, Z is =C(H)-, and Itx is pyridin-2-yl, 2,4-dichlorophenyl or 3-methylphenyl; then X3 is other than =C(CH3)-, =C(CH2CH3)-, or =C(cyclopropy1)-when X2 is =N-, XI is =C(CH3)-, and X3 is =C(CH3)-, the R" is other than 2,4-difluorophenyl or 3-chloro-4-cyanophenyl; and the compound is other than:
Ph rTh F N
I I N-N
___________________ C-101¨ j =
rlykl-s/
Ma , a 0 N ===""

µCr. fNH
¨

N H I i H
CI 1%1 ""'-µ2 N
0 , or 0 10011.31 In certain embodiments, the invention provides a compound of Formula 11:

N
R4a N N

R' a 0 (11), or a pharmaceutically acceptable salt thereof, wherein:
A is CH or N;
Rla is selected from -C1-C2 alkyl and -0-(C1-C2 alkyl), wherein RI' is optionally substituted with one or more fluoro;
R4a is selected from -(C1-C4 alkylene)-0-(CI-C3 alkyl), 1 -substituted-pipieridin-4-yl, C3-C6 cycloallcyl optionally substituted with one or more fluor , and tetrahydropyranyl; and R13 is selected from hydrogen, halo, phenyl, pyridinyl, and -0-(CI-C4 [001141 In some embodiments of Formula II, Rla is selected from -OCH3, -CH3, -OCHF2, and -CH2CH3.
[001151 In some embodiments of Formula IL R42 is selected from -CH200-13, -CH(CH3)0CH3, 4,4-difluorocyclohexyl, cyclopropyl, tetrayhyrdopyran-4-yl, 1 -(t-butoxycarbony1)-piperidin-4-yl, 1 -(isobutoxycarbony1)-piperidin-4-yl, 1-(isopropoxycarbony1)-piperidin-4-yl, 1 -(2-fluoroethyl)-piperidin-4-yl, 1 -(2,2-difl uoroethyl)-piperidin-4-yl, 1-(2,2,2-trifluoroethyl)-piperidin-4-yl, 1-(2-hydroxyisobuty1)-piperidin-4-yl, 1 -(hydroxyisopropylcarbony1)-piperidin-4-yl, 1 -(ethoxycarbonylmethyl)-piperidin-4-yl, 1 -(isopropylcarbony1)-piperidin-4-yl, 1 -methylpiperidin-4-yl, 1 -(methylsulfony1)-piperidin-4-y1 , 1 -(ethylsulfony1)-piperidin-4-y1 , I -(isopropylsul fony1)-piperidin-4-yl, 1 -(pheny1)-piperid in-4-yl, 1 -(oxetan-3-yl)piperidin-4-yl, 1-(pyridin-2-y1)-piperidin-4-yl, and 1-(pyrimidin-2-y1)-piperidin-4-Yi=
[001161 In some embodiments of Formula II, R13 is selected from hydrogen, chloro, fluor , -OCH(CH3)2, phenyl, and pyridin-2-yl.
[001171 Exemplary compounds of formula I and II are set forth in Figure 1. In some cases two (or more) of the compounds in Figure 1 having one (or more) wavy bonds will have the exact same structure. Because the wavy bond represents a chiral center of undetermined optical rotation, such compounds will be understood to be separate and distinct optical isomers of one another. Figure 1 is annotated to indicate those sets of two or more compounds that have the same depicted structure, but are of different stereochemistry.
4. Uses, Formulation and Administration Pharmaceutically acceptable compositions 1001181 According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this invention is such that is effective to measurably modulate a histone methyl modifying enzyme, or a mutant thereof, in a biological sample or in a patient.
In certain embodiments, the amount of compound in compositions of this invention is such that is effective to measurably modulate a histone methyl modifying enzyme, or a mutant thereof, in a biological sample or in a patient.
[001191 in certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition. in some embodiments, a composition of this invention is formulated for oral administration to a patient.
1001201 The term "patient," as used herein, means an animal, preferably a mammal, and most preferably a human.
[001211 The term "pharmaceutically acceptable carrier, adjuvant, or vehicle"
refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyffolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.

"pharmaceutically acceptable derivative" means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, cithcr directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
[001231 Compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
The term "parentcral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathccal, intrahepatie, intratesional and intracranial injection or infusion techniques. Preferably, the compositions arc administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
1001241 For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used TM TM
surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which arc commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
1001251 Pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral usc, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stcaratc, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[001261 Alternatively, pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
[001271 Pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[00128] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation.
Topically-transdermal patches may also be used.
1001291 For topical applications, provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
1001301 For ophthalmic use, provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonitnn chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.

[001311 Pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[001321 Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
[001331 The amount of compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon. the host treated and the particular mode of administration. Preferably, provided compositions should.
be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
[001341 It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions [001351 Compounds and compositions described herein are generally useful for the modulating of activity of one or more enzymes involved in epigenetic regulation.
[001361 Epigenetics is the study of heritable changes in gene expression caused by mechanisms other than changes in the underlying DNA sequence. Molecular mechanisms that play a role in epigenetic regulation include DNA methylation and chromatin/histone modifications. Histone methylation, in particular, is critical in many epigenetic phenomena.
[001371 Chromatin, the organized assemblage of nuclear DNA and histone proteins, is the basis for a multitude of vital nuclear processes including regulation of transcription, replication, DNA-damage repair and progression through the cell cycle. A number of factors, such as chromatin-modifying enzymes, have been identified that play an important role in maintaining the dynamic equilibrium of chromatin (Margueron, et al. (2005) Curr. Opin.
Genet. Dev. 15:163-176).
[001381 Histones are the chief protein components of chromatin. They act as spools around which DNA winds, and they play a role in gene regulation. There are a total of six classes of histones (H1, H2A, 112B, H3, 114, and 15) organized into two super classes:
core histones (H2A, H2B, H3, and H4) and linker histones (HI and H5). The basic unit of chromatin is the nucleosome, which consists of about 147 base pairs of DNA wrapped around the histone octamer, consisting of two copies each of the core histones H2A, H2B, H3, and 114 (Luger, et al.
(1997) Nature 389:251-260).
[00139] Histories, particularly residues of the amino termini of histones H3 and 114 and the amino and carboxyl termini of histones H2A, 112B and H1, are susceptible to a variety of post-translational modifications including acetylation, methylation, phosphorylation, ribosylation, sumoylation, ubiquitination, citrullination, deimination, and biotinylation.
The core of histones H2A and 113 can also be modified. Histone modifications are integral to diverse biological processes such as gene regulation, DNA repair, and chromosome condensation.
[00140j The present disclosure provides compounds and compositions for modulating activity of histone methyl modifying enzymes. Histone methyl modifying enzymes are key regulators of cellular and developmental processes. Histone methyl modifying enzymes may be characterized as either histone methyl transferases or histone demethylases. Histone demethylase enzymes have modules that mediate binding to methylated residues. For example, multiple demethylases contain a Tudor domain (e.g., IMID2C/GASCI) or a PHD domain (e.g., JAR1D1C/SMCX, PHF8).
1001411 The lysine specificities of many histone methyltransferases have been characterized.
For example SET7/9, SMYD3, and MLL1-5 are specific for H3K4. SUV39H1, DIM-5, and G9a are specific for H3K9. SET8 is specific for H4K20.
[00142] DOTI is an example of a non-SET domain containing histone methylase.
DOTI
methylates H3 on lysine 79.
[00143] Just as histone methylases have been shown to regulate transcriptional activity, chromatin structure, and gene silencing, demethylases have also been discovered which impact gene expression. LSD1 was the first histone lysine demethylase to be characterized. This enzyme displays homology to FAD-dependent amine oxidases and acts as a transcriptional corepressor of neuronal genes (Shi et al., Cell 119:941-953, 2004). Additional demethylases defining separate demethylase families have been discovered, including JHDM I
(or KDM2), JHDM2 (or KDM3), JMJD2 (or KDM4), JARID (or KDM5), JM.TD3 (or 1 DM6), and families (Lan et al., Curr. Opin. Cell Biol. 20(3):316-325, 2008).
1001441 Demethylases act on specific lysine residues within substrate sequences and discriminate between the degree of methylation present on a given residue. For example, LSD I
removes mono- or dimethyl- groups from H3K4. Members of the JARID1A-D family remove trimethyl groups from H3K4. UTX and JMJD3 demethylate H31(27, counteracting effects of EZH2 methylase activity. Substrate specificities of other demethylases have been characterized (see Shi, Nat. Rev. 8:829-833, 2007).
[001451 One class of histone methylases is characterized by the presence of a SET domain, named after proteins that share the domain, Su(var)3-9, enhancer of zeste [EVA, and trithorax.
A SET domain includes about 130 amino acids. SET domain-containing methylase families include SUV39H1, SET], SET2, EZH2, RJZ I, SMYD3, SUV4-20H1, SET7/9, and PR.-SET7ISET8 families (reviewed in Dillon et al., Genome Biol. 6:227, 2005).
Members of a family typically include similar sequence motifs in the vicinity of and within the SET domain.
The human genome encodes over 50 SET domain-containing histone protein methylases, any of which can be used in an assay described herein.
[001461 EZH2 is an example of a human SET-domain containing methylase. EZH2 associates with EED (Embryonic Ectoderm Development) and SUZ12 (suppressor of zeste 12 homolog) to form a complex known as PRC2 (Polycomb Group Repressive Complex 2) having the ability to tri-methylate histone H3 at lysine 27 (Cao and Zhang, Mol. Cell 15:57-67, 2004).
PRC2 complexes can also include RBAP46 and RBAP48 subunits.
[001471 The oncogenic activities of EZH2 have been shown by a number of studies. In cell line experiments, over-expression of EZH2 induces cell invasion, growth in soft agar, and motility while knockdown of EZH2 inhibits cell proliferation and cell invasion (Kleer et al., 2003, Proc. Nat. Acad. Sci. USA 100:11606-11611; Varambally et al., (2002), "The polycomb group protein EZH2 is involved in progression of prostate cancer," Nature 419, 624-629). It has been shown that EZH2 represses the expression of several tumor supressors, including E-cadherin , DAB2IP and RUNX3 among others. in xenograft models, EZH2 knockdown inhibits tumor growth and metastasis. Recently, it has been shown that down modulation of EZH2 in murine models blocks prostate cancer metastasis (Min et al., "An oncogene-tumor suppressor cascade drives metastatic prostate cancer by coordinately activating Ras and nuclear factor-kappaB," Nat Med. 2010 Mar; 16(3):286-94). EZH2 overexpression is associated with aggressiveness of certain cancers such as breast cancer (Kleer et al., Proc.
Nat. Acad. Sci. USA
100:11606-11611, 2003). Recent studies also suggest that prostate cancer specific oncogenic fusion gene TMPRSS2-ERG induces repressive epigenetic programs via direct activation of EZH2 (Yu et al., "An Integrated Network of Androgen Receptor, Polycomb, and ER.G Gene Fusions in Prostate Cancer Progression," Cancer Cell. 2010 May 18;17(5):443-454).
[001481 In some embodiments, compounds of the present invention modulate the activity of one or more enzymes involved in epigenetic regulation. In som.e embodiments, compounds of the present invention modulate the activity of a histone methyl modifying enzyme, or a mutant thereof. In some embodiments, compounds of the present invention modulate EZH2 activity. In some embodiments, compounds of the present invention down-regulate or suppress the activity of EZH2. In some embodiments, compounds of the present invention are antagonists of EZH2 activity.
[001491 In some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with a histone methyl modifying enzyme.
Accordingly, in some embodiments, the present invention provides a method of modulating a disease and/or disorder associated with a histone methyl modifying enzyme. In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with a histone methyl modifying enzyme comprising the step of administering a compound or composition of formula I.
1001501 in some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with overexpression of EZH2. in some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with overexpression of EZH2 comprising the step of administering a compound or composition of formula I. In some embodiments, the above method additionally comprises the preliminary step of determining if the subject is overexpressing EZH2.

[001511 In some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with cellular proliferation. In some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with misregulation of cell cycle or DNA
repair. In some embodiments, compounds and compositions of the present invention are useful in treating cancer. Exemplary types of cancer include breast cancer, prostate cancer, colon cancer, renal cell carcinoma, glioblastoma multiforme cancer, bladder cancer, melanoma, bronchial cancer, lymphoma and liver cancer.
[001521 The study of EZH2 deletions, rrtissense and frameshift mutations suggest that EZH2 functions as a tumor suppressor in blood disorders such as myelodysplastic syndromes (MDS) and myeloid malignancies (Ernst et al., Nat Genet. 2010 Aug; 42(8):722-6;
Nikoloski et al., Nat Genet. 2010 Aug; 42(8):665-7). Accordingly, in some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with the presence of a mutant form of EZH2. In some embodiments, compounds and compositions of the present invention are useful in treating diseases and/or disorders associated with the presence of Y641N EZH2. In some embodiment, the disease or disorder associated with the presence of a mutant form of EZ112 is a human .B cell lymphoma. In som.e embodiments, the disease and/or disorder associated with the presence of Y641N
EZF12 is follicular lymphoma or diffuse large-B-cell lymphoma. In some embodiments, compounds or compositions of the present invention are useful in treating blood disorders, such as myelodysplastic syndromes, leukemia, anemia and cytopenia. Sneeringer et al., "Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H31(27) in human B-cell lymphomas," Proceedings of the National Academy of Sciences, PNAS Early Edition published ahead of print on November 15, 2010.
1001531 In some embodiments, the present invention provides a method of reducing the activity of EZH2 in a subject comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of reducing the activity of wide-type EZH2 in a subject comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of reducing the activity of a mutant form of EZH2 in a subject comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of reducing the activity of a mutant form of EZH2 in a subject comprising the step of administering a compound or composition of formula I, wherein the mutant form of EZH2 is Y641N EZH2. In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with EZH2 comprising the step of administering a compound or composition of formula L In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with wide-type EZH2 comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with a mutant form of EZH2 comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with a mutant form of EZH2 comprising the step of administering a compound or composition of formula I, wherein the mutant form of EZH2 is Y641N EZH2. In some embodiments, the above method additionally comprises the preliminary step of determining if the subject is expressing a mutant form of EZH2, such as Y641N EZH2.
In some embodiments, the present invention provides a method of reducing the activity of a mutant form of EZH2, such as Y641N EZEI2, in a subject in need thereof comprising the step of administering a compound or composition of formula I. In some embodiments, the present invention provides a method of treating a subject suffering from a disease and/or disorder associated with a mutant form. of EZH2 comprising the step of administering a compound or composition of formula I. In some embodiments, the above method additionally comprises the preliminary step of determining if the subject is expressing a mutant form of EZH2, such as Y641N EZH2. In some embodiments, that determination is made by determining if the subject has increased levels of histone H3 Lys-27-specific trimethylation (H3K27ine3), as compared to a subject known not to express a mutant form of EZH2.
EQUIVALENTS
1.00154j The representative examples that follow are intended to help illustrate the invention, and are not intended to, nor should they be construed to, limit the scope of the invention. Indeed, various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including the examples that follow and the references to the scientific and patent literature cited herein.
1001551 It will be appreciated that for compound preparations described herein, when reverse phase HPLC is used to purify a compound, a compound may exist as an acid addition salt. In some embodiments, a compound may exist as a formic acid or mono-, di-, or tri-trifluoroacctic acid salt.
1001561 It will further be appreciated that the present invention contemplates individual compounds described herein. Where individual compounds exemplified arc isolated and/or characterized as a salt, for example, as a trifluoroacetic acid salt, the present invention contemplates a free base of the salt, as well as other pharmaceutically acceptable salts of the free base.
1001571 The following examples contain important additional information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and the equivalents thereof EXAMPLES
1001581 As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the synthetic methods and Schemes depict the synthesis of certain compounds of the present invention, the following methods and other methods known to one of ordinary skill in the art can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
1001591 Unless otherwise noted, all solvents, chemicals, and reagents were obtained commercially and used without purification. The 11:1 NMR spectra were obtained in CDCI3, 1/6-DMSO, CD30D, or (16-acetone at 25 C at 300 MHz on an OXFORD (Varian) with chemical shift (6, ppm) reported relative to TMS as an internal standard. HPLC-MS
chromatograms and spectra were obtained with Shimadzu LC-MS-2020 system. Chiral analysis and purification were obtained with Yilite P270.
1001601 Example 1. Synthesis of N4(4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-v1)methyl)-5-methyl-1-phenyl-1H-pyrazole-4-carboxamide (Compound 100).

, H
H \ CySN'N0 0 0 0 [00161.1 A mixture of 3-(aminomethyl)-4,6-dimethylpyridin-2(11.0-one (70 mg, 0.46 mmol), 5-methyl-I -pheny1-1H-pyrazole-4-earboxylic acid (93 mg, 0.46 miriol), 0-(7-azaberizotriazole-1-y1)-N,N,N',N4etramethyluronium. hexafluorophosphate (210 mg, 0.55 mmol) and triethylamine (70 mg, 0.69 mmol) in anhydrous dichloromethane (5 mi.) was stirred at room temperature for 15 hours. Then the mixture was filtered and the solid was washed with water (10 mL), methanol (10 mL) and dichloromethane (10 mL) in turns to give N44,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yOmethyl)-5-methyl-1-phenyl-lH-pyrazole-4-carboxamide as a white solid (40 mg, 26%). LRMS (M H.1) m/z: calcd 336.16; found 336.
1001621 .Example 2., Synthesis of N4(4,6-dimethyl-2-oxo-1,2-dillydroovridin-3-ynniethy14-1-(ohenyisu 1 fon yi)-11-1-i zi ie-3-carboxami e (Compou n (i I
,15).
c) 0, 0,r Ni.i 2 + FlOyike\
HN

[001631 A mixture of 3-(aminomethyl.)-4,6-dimethylpyridin-2(111)-one (100 mg, 0.65 mmol.), 1-(phenylsulfony1)-1H-indole-3-carboxylic acid (196 mg, 0.65 mmol), 047-azabenzotriazole-1-y1)-NAU,N'tetrarnethyluronium hexafluorophosphate (319 mg, 0.84 mniol) and triethylamine (98 mg, 0.97 mmol) in anhydrous dichloromethane (10 mL) was stirred at room temperature for 15 hours. Then the mixture was filtered and the solid was washed with water (10 mL), methanol (10 mL) and dichloromethane (10 mL) in turns to give N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-1-(phenylsulfony1)-1H-indole-3-carboxamide as a white solid (78 mg, 28%). L-RMS (M calcd. 435.13; found 435.
[001641 Example 3., Synthesis of (R or S)-N4(2-h roiv-4,6-dimethvirevridin-3-vbmethvb-3-methyl-1-(1-phenviethvi}-11/-Dyrazole-4-earhoµ amide (Com)ound 106) and (R or S)-N((2-Irs d rox 1-pyridin-3-vhout I)-3-met It I-1-(1-ohenvlethvl)-1H-pvrazole-4-carboKamide (Compound I

CO2HcQMBUCH
TFe0.4/-:
SOC,2 Me0H ei\rõ= KjeOj DtVE ==itep 2 meo2c I IF
=== 1102C I
Step 1 Hig-N StepN 3 bo HOB T. LX:I cv.:-:!,roar:x.t;
OH 0 <r) 1.
.3.. D., OH Q
f Step 4 Step 5 * titti " = )'`'N
1001651 Synthesis of methyl 3-methyl4H-pyrazole-4-carboxylate.
CO 2H Cape SOCl2, Me011 __________________________________________ vi HN-N Step 1 HN-N
1001661 To a solution of 3-methyl-1H-pyrazole-4-carboxylic acid (1.26 g, 10 mmol) in methanol (100 mL) was added thionyl chloride (5.73 g, 48 mmol) at 0 C. The mixture was stirred for 12 hours. The solvent was evaporated in vacuo. To the residue, saturated sodium bicarbonate aqueous solution was added and the mixture was extracted with ethyl acetate (100 mLx 3). The organic phases were combined, dried over sodium sulfate, filtered and concentrated in VaCUO to give methyl 3-methy1-1H-pyrazole-4-carboxylate (0.8 g, 57%). 111 NMR (300 MHz, CDCI3): 6 7.87 (s, I H), 3.84 (s, 3H), 2.53 (s, 3H).
[001671 Synthesis of methyl 3-methy1-1-(1-phenylethyl)-1H-pyrazole-4-carboxylate and methyl 5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylate.
CO2Me (cr._ Br * K2CO3, Me02C.t.X. 440 / Step 2 HN-NN,N' 1001681 To a solution of methyl 3-methyl-1H-pyrazole-4-carboxylate (280 mg, 2 mmol) in N, N-dimethylformamide (30 mL) was added (1-bromoethyl) benzene (0.37 g, 2 mmol) and potassium carbonate (0.55 g, 4 mmol). The mixture was stirred at 20 C and stirred for 12 hours.
The solvent was evaporated in vacuo and the residue was purified by CXTH
(Colurrm:Dsisol, 10 1.tM, C18, 250 rnm*50 mm; Mobile: acetonitrile(0.1% formic acid)-water (0.1%
formic acid), acetonitrile from 30% to 70% in 80 minutes; oven: 20 C; flow rate: 50 mliminute, wavelength:
214 mu) to give methyl 3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylate (90 mg, 19%) and methyl 5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylate (80 mg, 16%).
The product was used directly in the following reaction. LRMS (M H') m/z: calcd 244.12;
found 244.
1001691 Synthesis of 3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylic acid.

LiOH
THF/Me0H/H20 Me02C¨bli HO2C-6 so]
Step 3 1001701 A mixture of methyl 3-methyl-I -(1-phenylethyl)-1H-pyrazole-4-carboxylate (90 mg, 0.37 mmol), lithium hydroxide monohydrate (57.1 mg, 1.36 mmol), tetrahydrofuran (5 mL), methanol (1mL) and water(' mL) was stirred at 20 C for 4 hours. The mixture was acidified to pH=1 with concentrated hydrochloric acid and then extracted with ethyl acetate (15 mL x 3).
The combined organic phase was dried by sodium sulfate, and then filtered. The filtrate was concentrated in vacuo to give 3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylic acid as a white solid (60 mg, 70%). 111 NMR (300 MHz, d-DMS0): ö 12.24 (s, 1H), 7.82 (s, 1H), 7.36-
7.17 (m, 5H), 5.67 (q, J= 7.2 Hz, 1H), 2.43 (s, 3H), 1.79 (d, J = 6.9 Hz, N).
1001711 Synthesis of N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide.

HOBT, EDCI OH 0 HO2C¨b +
Et3N, DCM
I H
Step 4 \ -1%1 1001721 A mixture of 3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylic acid (60 mg, 0.26 mmol), 1-ethy1-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (95 mg, 0.5 mmol), N-hydroxybenzotrizole (67 mg, 0.5 mmol), triethylamine (0.1 mL) and dichloromethane (5 mL) was stirred at 25 C for 0.5 hours. And then 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (50 mg, 0.33 mmol) was added. The mixture was stirred at 25 C for 12 hours. To the mixture, water (10 ml) was added and the mixture was extracted with dichloromethane (10 mL x 3).
The combined organic phase was dried by sodium sulfate and then filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica gel, dichloromethane /methanol = 20:1) to give N-((2-hydroxy-4,6-dimethylpyridin-3-yOmethyl)-3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide as a white solid (56 mg, 58%). LRMS (M
+ m/z:
calcd 364.19; found 364. 1H NMR (300 MHz, d5-DMS0): h 11.46 (s, 1H), 7.96(s, 1H), 7.82 (t, = 4.5 Hz, 1H), 7.33-7.13 (m, 5H), 5.84 (s, 1H), 5.62 (q, J = 7.2 Hz, 1H), 4.20 (dõ/.= 5.1 Hz, 2H), 2.40 (s, 3H), 2.14 (s, 3H), 1.99 (s, 3H), 1.76 (d, .1= 7.2 Hz, 3H).
[00173] Synthesis of (R or S)-N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide (Compound 106) and (R or hydroxy-4,6-dhnethylpyridin-3-Amethyl)-3-methyl-1-(1-phenylethyl)-1H-pyrazole-carboxamide (Compound 105).
Chira OH 0 * separl HPLCation OH 0 = OH 0 .5 X Step 5 * N , I H
1001741 N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-3-methyl-1-(1-phenylethyl)-11-1-pyrazole-4-carboxamide was separated by chiral HPLC (condition: column AD-H
(20mm*250rnm*51.1.m), hexane: ethanol (0.2% DEA) 50: 50, flow rate: 13 ml/mm).
The two isomers of N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-3-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide were obtained, but absolute stereochemistry was not determined. The retention times were 9.552 minutes ("Peak 1"; Compund 105) and 15.505 minutes ("Peak 2";
Compound 106) in chiral HPLC chromatography. LRMS (M + H') calcd. 364.19;
found 364. 1H NMR (300 MHz, ti6-DMS0): 6 11.46 (s, 1H), 7.96 (s, 1H), 7.82 (t, J =
4.5 Hz, 1H), 7.33-7.13 (m, 5H), 5.84 (s, 1H), 5.62 (q, J= 7.2 Hz, 1H), 4.20 (d, J = 5.1 Hz, 2H), 2.40 (s, 3H), 2.14 (s, 3H), 1.99 (s, 3H), 1.76 (d, J = 7.2 Hz, 3H).
1001751 Example 4. Synthesis of (R or S)-N4(2-hydroxv-4,6-dimethylpyridin-3-v1)methyl)-5-methvl-1-(1-phenviethvi)-11/-pvrazole-4-carboxamide (Compound 107) and (R or SI-N-((2-hydroxy-4-dimethi, 1-pyridin-3-1-1)methyl)-5-methyl-1-(1-phenylethvg-1H-pyrazole-4-ca rboxa mid e (Compound 108 LION HOU, EDO
Me02C, ¨ THF)Me0H/1120 HO2C.r. + t.1 EI3N. CCM
7 II¨

Stp 1 Step 2 CH 0 Chiral HPLC dx \
separation )CCifjty,N
Step 3 z [001761 Synthesis of 5-methyl-1-(1-phenylethyl)-11/-pyrazole-4-carboxylic acid.
mo2c,r, 4111 = LiOH
Me02C,r, THF/Me0H/H20 N
Step 1 1100177j A mixture of methyl 5-methyl-I -(1-phenylethyl)-1H-mazole-4-carboxylate (80 mg, 0.32 mmol), lithium hydroxide monohydrate (57.1 mg, 1.36 mmol), tetrahydrofuran (5 mL), methanol (1mL) and water (1 mL ) was stirred at 20 'C for 4 hours. The mixture was acidified to pH=1 with concentrated hydrochloric acid and then extracted with ethyl acetate (15 mL x 3).
The combined organic phase was dried by sodium sulfate, and then filtered. The filtrate was concentrated in vacuo to give 5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylic acid as a white solid (49 mg, 62%). 11-1 NMR (300 MHz, ce-DMS0): 6 12.16 (s, 11), 8.26 (s, 1H), 7.37-7.25 (m, 5H), 5.55 (q, J = 7.2 Hz, 1H), 2.30 (s, 3H), 1.78 (d, J = 6.9 Hz, 3H).
[001781 Synthesis of N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide.

HOB?. EDO!
= e HO2C NH2 Et3N, 0CM )Lr%
N = "`..=== y: Step 2 OH
(00179) A mixture of 5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxylic acid (49 mg, 0.21 mmol), 1-ethy1-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (95 mg, 0.5 mmol), N-hydroxybenzotrizole (67 mg, 0.5 mmol), triethylamine (0.1 mL) and dichloromethane (5 mL) was stirred at 25 C for 0.5 hours. Then 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (50 mg, 0.33 mmol) was added. The mixture was stirred at 25 C for 12 hours. To the mixture, water (10 ml) was added and the mixture was extracted with dichloromethane (10 mi. x 3). The combined organic phase was dried by sodium sulfate and then filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica gel, dichl.oromethane /methanol = 20:1) to give N4(2-hydroxy-4,6-di methylpyridin-3-yOmethyl)-5-m ethyl-141-phenylethyl)-1H-pyrazole-4-carboxamide as a white solid (43 mg, 56%). LRMS (M
W) m/z:
calcd 364.19; found 364. III NMR (300 MHz, d6-DMS0): 6 11.46 (s, 1H), 8.34(s, 1H), 7.70 (t, I
= 5.1 Hz, 11-1), 7.35-7.18 (m, 5H), 5.84 (s, 1H), 5.49 (q, J= 7.2 Hz, 1H), 4.20 (d, .1" 4.8 Flz, 2H), 2.29 (s, 31-1), 2.15 (s, 3H), 2.10 (s, 3H), 1.73 (d, J --= 6.9 Hz, 311).
(001801 Synthesis of (R or S)-N-((2-hydroxy-4,6-dimethylpyridin-3-yOmethyl)-5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide (Compound 107) and (R or S)-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-5-methyl-1-(1-phenylethyl)-1H-pyrazole-4-carboxamide (Compound 108).

ot-i 0 OH 0 OH 0 Chiral HPLC
N riLliksp separation N".
.......................... 41.
-N
Step 3 1001811 AT-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-5-methyl-1-(1-phenylethyl)-1H-pyrazol e-4-carboxamide was separated by chiral HPI,C (condition; column AD-H
(20mm*250rnm*51,tm), hexane: ethanol (0.2% DEA) = 50: 50, flow rate: 13 ml/min). The two isomers of AT-((2-hydrox y-4 ,6-dirnethyl.py rid in-3-yl)methyl)-5-methyl-1-(1-phenyl.ethyl)-1H-pyrazole-4-carboxamide were obtained, but absolute ste,reochemistry was not determined. The retention times were 6.574 minutes ("Peak 1"; Compound 107) and 7.974 minutes by Chiral HPLC chromatography ("Peak. 2"; Compound 108). LRMS (M H m/z: calcd 364.19;
found 364. 111 NMR. (300 MHz, d6-DMS0): 6 11.46 (s, 111), 8.34 (s, 1.11), 7.70 (t, J= 5.1 Hz, 1.FI), 7.35-7.18 (m., 5H), 5.84 (s, Ill), 5.49 (q, J = 7.2 Hz, U), 4.20 (d, J = 4.8 Hz, 2H), 2.29 (s, 3H), 2.15 (s, 3H), 2.10 (s, 3H), 1.73 (d, ./..= 6.9 Hz, 3H).
tool 821 Example 5., Synthesis of N-02-hydroxv-4.6-dimethvinyridin-3-y1)methyl)-5-methyl-1-(1-beniVO-1/1-0Vrazole-4-carboxamide (Compound 109).
tos02C UOH
SOC. Me0H JrIV:03. OW THF/11/1e0H/H20 Step 1 - Ey step 2 vi=N I Step 3 C0711 602Me HOST, MCI OH 0 HO2C4-11".0 Uelsi N
-.1k1 Nikõ..14H2 Step 4 OH
[00183j Synthesis of methyl 3-methyl-1.ff-pyrazole-4-earboxylate.
N-NH N-NH
Ase SOCl2, Me0H
Step CO2H CO2Me [001841 To a solution of 3-methyl-1H-pyrazole-4-carboxylic acid (1.26 g, 10 mmol) in methanol (100 mL) was added thionyl chloride (5.73 g, 45 mmol) at 0 C. The mixture was stirred for 12 hours. The solvent was evaporated in vacuo. To the residue, saturated sodium bicarbonate aqueous solution was added and the mixture was extracted with ethyl acetate (100 mi,x 3), the organic phase was dried by sodium. sulfate. The mixture was filtered and the filtrate was concentrated in yam to give methyl 3-methyl-1H-pyrazole-4-carboxyl.ate (0.8 g, 57%). 11-1 NMR (300 MHz, CDC13): 6 7.87 (s, 1H), 3.84 (s, 3H), 2.53 (s, 3H).

[00185] Synthesis of methyl 1-benzy1-5-methyl-1H-pyrazole-41-carboxylate.
N¨NH Me020, K2CO3, DMF
/
Br Step 2 CO2Me [001861 To a solution of methyl 3-methyl-111-pyrazole-4-carboxylate (280 mg, 2 minor) in N, N-dimethylformamide (30 mL) was added (bromomethyl)benzene (0.34 g, 2 mmol) and potassium carbonate (0.55 g, 4 mmol). The mixture was stirred at 20 C and stirred for 12 hours.
The solvent was evaporated in vacuo and the residue was purified by cxm (Column:Dsisol, 101.tM, C18, 250 rtun*50 mm; Mobile: acetonitrile(0.1% formic acid)-water (0.1% formic acid), acetonitrile from 30% to 70% in 80 minutes; oven: 20 C; flow rate: 50 ml/minute, wavelength:
214 urn) to give crude methyl 1-benzy1-5-methy1-1H-pyrazole-4-carboxylate (200 mg, 42%).
LRMS (M + Er.) m/z: calcd. 230.11; found 230.
[00187] Synthesis of 1-benzy1-5-methyl-1H-pyrazo rboxyl it; acid.
Me02C LOH
INF/MOH/HP HO2 C N, 4.-.1/ Step 3 ¨N
N -(001881 A mixture of methyl 1-benzy1-5-methy1-1H-pyrazole-4-carboxylate (200 mg, 0.86 mmol), lithium hydroxide monohydrate (Ill mg, 2.62mmo1), tetrahydrofuran (10 ittL ), methanol (2 mL ) and water(2 mL ) was stirred at 20 C for 4 hours. The mixture was acidified to p11=1 with concentrated hydrochloric acid and then extracted with ethyl acetate (15 mL x 3).
The combined organic phase was dried by sodium sulfate, and then filtered. The filtrate was concentrated in vacuo and separated by chiral HPLC (condition: column AD-H
(20mm*250mm*511m), hexane: ethanol (0.2% DEA) = 50: 50, flow rate: 13 rni.imin) to give 1-benzy1-5-methyl-lif-pyrazole-4-carboxylic acid (75 mg, 40%). LRMS (M + 1.14) m/z: calcd.
216.09; found 216.
[00189] Synthesis of 1-benzyl-N42-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-5-methyl-1H-pyrazole-4-carbox amide (Compound 109).

H024;4'-µ0 Et3N, ¨4 I I
DCM I H .14 N NN2 Step 'N
\-"{) [001901 A mixture of 1-benzy1-5-methyl-1H-pyrazole-4-carboxylic acid (75 mg, 0.34 mmol), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (95 mg, 0.5 mmol), N-hydroxybenzotrizole (67 mg, 0.5 rnmol), triethylamine (0.1 mL) and dichloromethane (5 mL) were stirred at 25 C for 0.5 hours. Then 3-(aminomethy1)-4,6-dimethylpyridin-2-ol (50 mg, 0.33 mmol) was added. The mixture was stirred at 25 C for 12 hours. To the mixture, water (10 nil) was added and the mixture was extracted with dichloromethane (10 mL x 3).
The combined organic phase was dried by sodium sulfate and then filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica gel, dichloromethane /methanol = 20:1) to give 1-benzyl-N-((2-hydroxy-4,6-dimethylpyridin-3-yOmethyl)-5-methyl-IH-pyrazole-4-carboxamide as a white solid (35 mg, 29%). LRMS (M + Fr) nilz:
calcd 350.17;
found 350. '11 NMR (300 MHz, d6-DMS0): ô 11.46 (s, 111), 8.21 (s, 1.H), 7.68 (t, J= 5.1 Hz, 111), 7.38-7.21 (m, 5H), 5.85 (s, 111), 5.20 (s, 2H), 4.19 (d, J= 5.4 Hz, 211), 2.29 (s, 311), 2.15 (s, 3H), 2.10 (s, 3H).
[001911 Example 6. Synthesis of N-((2-hydroxv-44-ditnethyliovridin-3-vDmethyl)-I-1 H-p razole-4-carboxamide (Compound 11 M.
LiOH
N¨NHfi K2CO3, DMF Me02C * THF/Me0H/H20 CO2Me methyl 5-methyl-I -phenethy1-1H-pyrazole-4-carboxylate HOBT, EDCI
= Et:sN. DCM N / CO2H 'NT = HO f--NH
N NH2 Step 3 NO¨ =
[001921 Synthesis of methyl 5-methyl-1.-phenethy1-111-pyrazole-4-carboxylate.
N¨NH Me02C
K2CO3, DMF
Br Step 1 CO2Me [001931 To a solution of methyl 3-methyl-1H-pyrazole-4-carboxylate (280 mg, 2 mrnol) in .N,N-dimethylformamide (30 mL) was added (bromoethyl)benzene (0.37 g, 2 mm.ol) and potassium carbonate (0.55 g, 4 mmol). The mixture was stirred at 20 C for 12 hours. The solvent was evaporated in vacuo and the residue was purified by CXTH
(Column:Dsisol, 10gM, C18, 250 mm*50 mm.; Mobile: acetonitrile (0.1% formic acid) - water (0.1%
formic acid), acetonitrile from 30% to 70% in 80 minutes; oven: 20 C; flow rate: 50 mUminute, wavelength:
214 nm) to give crude methyl 5-methyl-1-phenethyl-IH-pyrazole-4-carboxylate (177 mg, 36%).
The product was used for the next step directly. LRMS (M + Fr) m/z: calcd.
244.12; found 244.
1001941 Synthesis of 5-methyl-1-phenethy1-1.1/-pyrazole-4-carboxylic acid.
UOH
itie02c,e, THF/MeOH/H20 co2H
Step 2 401 [00195] A mixture of methyl 5-methyl-1-phenethyl-IH-pyrazole-4-carboxylate (177 mg, 0.72 mmol), lithium hydroxide monohydrate (111 mg, 2.62 mmol), tetrahydrofuran (5 mL ), methanol (1 mL ) and water(1 mL ) was stirred at 20 C for 4 hours. The mixture was acidified to pH=1 with concentrated hydrochloric acid and then extracted with ethyl acetate (15 mL x 3). The combined organic phase was dried by sodium sulfate, and then filtered. The filtrate was concentrated in vacuum and separated by HI'LC (condition: column AD-H (20 mm *
250 mm *
um), hexane: ethanol (0.2% DEA) = 50: 50, flow rate: 13 mL/min) to give 5-methyl-I -phenethy1-1H-pyrazolc-4-carboxylic acid (60 mg, 36%). LRMS (M + fl.+) m/z:
calcd. 230.11;
found 230.
[00196] Synthesis of N4(2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-5-methyl-1-phenethyl-IH-pyrazole-4-carboxatnide (Compound 110).

HOBT.
ft./ C 02 H Et3N, DCM N
40 sie, 3 N/
OH
[00197] A mixture of 5-methyl-I -phenethy1-1H-pyrazole-4-carboxylic acid (60 mg, 0.27 mmol), 1-ethyl-3-(3-dimethyl.aminopropyl.)carbodiimide hydrochloride (95 mg, 0.5 mmol), N-hydroxybenzotrizole (67 mg, 0.5 mmol), triethylamine (0.1 miL) and dichlorom.ethane (5 mL) were stirred at 25 C for 0.5 hours. Then 3-(arninornethyl)-4,6-dimethylpyridin-2-ol (50 mg, 0.33 mmol) was added. The mixture was stirred at 25 `-)C for 12 hours. To the mixture, water (10 ml) was added and the mixture was extracted with dichloromethane (10 mL x 3).
The combined organic phase was dried by sodium sulfate and then filtered. The filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica gel, dichl.oromethane /methanol = 20:1) to give N42-hydroxy-4,6-dimethylpyridin-3-yl)m.ethyl)-5-m.ethy1-1-phenethyl-IH-pyrazole-4-carboxamide as a white solid (30 mg, 31%). LRMS (M +
H4) m/z:

calcd 364.19; found 364. 11-1 NMR (300 MHz, d'-DMS0): 11.47 (s, 1H), 8.08 (s, 1H), 7.57 (t, .1 .= 5.1 Hz, 1H), 7.30-7.15 (in, 5H), 5.85 (s, 1H), 4.23-4.18 (m, 4H), 3.06 (t, .1 = 7.2 Hz, 2H), 2.31 (s, 3H), 2.15 (s, 3H), 2.11 (s, 3H).
1001981 Example 7. Synthesis of N4(2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-2-methyl-1-(1-phenylethyl) 1H-indole-3-carboxamide (Compound 111). (R or 51-N-((2-hydroxy-4,6-dimethylnyridin-3-yl)methyl) 2-methyl-1-1-phenylethyb 1H-Indole-3-carboxamide (Compound 1371 and (S or 1)-N-((2-hydroxy-4.6-diniethyloyridin-3-v1)methyl) 2-methyl-1-1-phenylethyl) 1H-indole-3-carboxamide (Compound 136).
Cu2O, cs2co3, frk.,r-d o o omsorNo love NeH, DMF, rt, e step 2 _ step 1 *
Br OH
0 HOST, EVCI, KOH, WOK reflux filk NH2 Et3N, DCM, rt reep3 41Ir N step 4 ¨
* N
1001991 Synthesis of ethyl 2-methyl-1H-indole-3-carboxylate.
110 o o Cu2O, cs2c03, cmsoki2o. loo*c_ NH2 ki step 1 IS N
[002001 To a mixed solution of dimethyl sulfoxide and water (20 2-iodobenzenamine (3.0 g, 13,7 mmol), ethyl 3-oxobutanoate (2.0 g, 15.1 mmol), copper(l) oxide (0.2 g, 1.4 mmol) and cesium carbonate (4.5 g, 13.7 mmol) was added. The mixture was stirred at 100 'C for 9 TM
hours under nitrogen gas atmosphere. The reaction mixture was filtered through a pad of celite.
The filtrate was diluted with water and extracted with ethyl acetate. The organic phase was concentrated in vacua, and then the residue was purified by column chromatography (silica gel, petroleum ether / ethyl acetate =5:1) to give ethyl 2-methy1-1H-indole-3-carboxylatc as a light yellow solid (0.42 g, 15%). ill NMR (300 MHz, CDC13): (5 8.12-8.09 (m, 1H), 7.31-7.16 (m, 311), 4.40 (q, J= 6.9 Hz, 2H), 2.77 (s, 3H), 1.45 (t, 6.9 Hz, 3H).
[00201 j Synthesis of ethyl 2-methyl-1-(1-nhenylethyl)-1H-indole-3-carboxylate.

Br NeH, DMF. rt step 2 \
N
[002021 A mixture of ethyl 2-methyl-1H-indole-3-carboxylate (400 mg, 1.97 mmol) and sodium hydride (47 mg, 2.0 mm.ol) in NN-dimethylformamide (5.0 mL) was stirred at room temperature for 0.5 hours, and then (1-bromoethypbenzene (361 mg, 2.0 mm.ol) was added. The mixture was stirred at room temperature for 3 hours. The reaction mixture was poured into ice-cold water and extracted with ethyl acetate. Organic layers were combined and concentrated to give a residue. The residue was purified by chromatography (petroleum ether /
ethyl acetate =
5:1) to give ethyl 2-methy1-1-(1-phenylethyl)-1/1-indole-3-carboxylate (340 mg, 56%). LRMS
(M +H.') m/z: calcd 307.16; found 307.
1002031 Synthesis of 2-ntethy1-1-(1-pheny1ethy)-1H-indole-3-carboxy1ic acid.

KOH, Me0H, reflux so step3 'N
;s0 1002041 To a mixed solution of methanol and water (4 mL), ethyl 2-methy1-1-(1-phenylethyl)-1H-indole-3-carboxylate (3(X) mg, 0.98 mtnol) and potassium hydroxide (546 mg, 9.76 mmol) was added. The mixture was refluxed with stirring for 4 hours. The reaction mixture was concentrated to give a residue. To the residue, water (10 mL) was added and the mixture was extracted with dichloromethane (20 mL x 3). T he organic phase was concentrated to give crude product 2-methyl-1-(1-phenylethyl)-1H-indole-3-carboxylic acid (150 mg). LRMS
(M-1-1)" m/z:
calcd 279.13; found 279.
1002051 Synthesis of N-((2-hydroxy-41.6-dimethylpyridin-3-Amethyl)-2-methyl-1-(1-phenylethyl)-1H-indole-3-carboxamide (Compound 114 OH
HO OH
0 HOBT, EDCI, ifµl \
igh \ 4. N -Th NH2 Et3N.rt , _ 1_,N
WA N
IP
1002061 To a solution of 2-methyl-I -(1-phenylethyl)- I H-indole-3-carboxylic acid (150 mg, 0.54 mmol) in anhydrous dichloromethane (10 mL) was added N-hydroxybenzotriazole (87 mg, 0.64 mmol), 1-ethyl-3[3-(dimethylamino)propyllearbodiimide hydrochloride (124 mg, 0.64 mmol) and trimethylamine (163 mg, 1.61 rrunol). The mixture was stirred at room temperature for 0.5 hours, then 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (98 mg, 0.64 nunol) was added.
The mixture was stirred at room temperature for 12 hours. To the reaction mixture was added water (20 mL), extracted with dichloromethane (20 mi., x 2). The organic layers were combined and concentrated to give a residue. The residue was purified by column chromatography(silica gel, dichloromethanelmethano1=20:1) to give compound N42-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-111-indole-3-carboxamide as an off-white solid (130 mg, 59%). LRMS (M H+) nez: calcd 413.21; found 413. 111 NIV1R (300 MHz, d6-DMS0) 6: 11.59 (s, IH), 7.73-7.66 (m, 211), 7.34-7.29 (m, 31-1), 7.23 (d, J= 7.5 Hz, 1H), 7.16-6.89 (in, 411), 5.94 Oh ../ = 7.2 Hz, 111), 5.88 (s, 1H), 4.32 (d, J = 5.4 Hz, 2H), 2.60 (s, 311), 2.27 (s, 311), 2.11(s, 311), 1.88 (d, J = 7.2 Hz, 311).
[002071 Synthesis or (R or 19-N-((2-hydroxy-4,6-dimethylpyridin-3-y1)methyl) 2-methyl-1-1-phenylethyl) 1H-indole-3-carboxamide (Compound 137) and (R or S)-N-((2-hydroxy-4,6-dim ethylpy rid in-3-y1)methyl) 2-methyl-1-1.-phenylethyl) 1H-indole-3-carboxamide (Corn p o u nd 136).
OH ps Chrial HMO 41.1....µ -0--- \ p o 41`ti_co . -..4 \ Hõ...{ n i4...eL11 i N,,"-.2 I
1002081 N42-hydroxy-4,6-dimethylpyridin-3-yl)methy1)2-methyl-1-1-phenylethy1)1H-indole-3-carboxamide (130 mg, 0.31 mmol) was separated by chiral prep-HPLC
(Daicel AD-H
(250 mm x 20 mm x 5 um), hexane: ethanol (0.2% DEA) = 50: 50, flow rate: 13 mL/min), then (R or 5) N -((2-hydroxy-4,6-dimethylpyridin-3-yl)methy I) 2-methy1-1-1-phenylethyl) 1H-indole-3-carboxamide (30 mg, 23%) and (S or R) N((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl) 2-methy1-1-1-phenylethyl) 1H-indole-3-carboxamide (30 mg, 23%) was obtained. The retention times were 8.030 minutes ("Peak 1"; Compound 137) and 14.126 minutes ("Peak 2"; Compound 136) respectively in chiral HPLC chromatography. LRMS (M+H-)m/2: calcd 413.21;
found 413.1H NMR (300 MHz, d6-DMS0) 6: 11.59 (s, 1H), 7.73-7.66 (m, 2H), 7.34-7.29 (m, 31.), 7.23 (d, J= 7.5 Hz, 1H), 7.16-6.89 (m, 4H), 5.94 (q, J= 7.2 Hz, 1H), 5.88 (s, 1H), 4.32 (d, J=
5.4 Hz, 2H), 2.60 (s, 3H), 2.27 (s, 3H), 2.11(s, 3H), 1.88 (d, J= 7.2 Hz, 3H).
[002091 Example 8. Synthesis of N4(2-hydroxy-4.6-dimethylpyridin-3-ybrneilty1)-1-(1-phenvlethyl)-1.H-indazole-3-carboxamide (Compound 1121.
00H CO2Me Br CO,Me COAle io ,N SOCl2, Me0H 4,14 K2C.02:

Step 1 N Step 2 UOH CO2H NH2 OH HOlitt 0 H Et2N, DCM tk?))---TEZ2,0 N
Step 4 I
[002101 Synthesis of methyl 1H-indazole-3-ea rboxy late.
COOH CO2Me 101 N SOCl2, Me0H
"N
Step 1 [002111 To a solution of 1H-indazole-3-carboxylic acid (5.0 g, 30.8 mmol) in methanol (50 mL), thionyl chloride (15 mL) was added dropwise at 0 C. After the addition, the mixture was heated to reflux and maintained at the temperature for 1.5 hours. Then the reaction mixture was concentrated to give a residue. To the residue was added saturated sodium bicarbonate (50 mL), and then extracted with ethyl acetate (50 mL x 3). The organic phase was combined and dried over anhydrous sodium sulfate. The mixture was filtered and the filtrate was concentrated under reduced pressure to give methyl 1H-indazole-3-carboxylate as a white solid (5.1 g, 94%). 1H
NMR (300 MHz, d-DMS0): (5 13.91 (s, 1H), 8.06 (d, 1= 8.2 Hz, 1H), 7.65 (d, J=
8.4 Hz, 1H), 7.44 (ddd, J= 8.3 Hz, J= 6.9 Hz, J=1.1 Hz, 1H), 7.30 (ddd, J= 7.9 Hz, J= 6.9 Hz, J= 0.9 Hz, 1H), 3.92 (s, 3H).
[002121 Synthesis of methyl 1-(1-phenylethyI)-1H-indazole-3-c-Arboxylate.

Me CO2Me Br CO2 CO2Me ",N
K2CO3, Acetone io Step 2 NIµ'N 1411 [00213] To a solution of methyl 1H-indazole-3-carboxylate (2.0 g, 11.4 mmol) in acetone (50 mL), were added potassium carbonate (1.6 g, 11.4 mmol) and (1-bromoethyl) benzene (2.2 g, 11.9 mmol). Then the mixture was refiuxed for 12 hours. The reaction mixture was concentrated to give a residue. To the residue was added water (50 mL), extracted with dichloromethane (50 mL x 3). The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
The residue was purified by column chromatography (silica gel, petroleum ether / ethyl acetate =5:1) to afford two isomers.
Methyl 1-(1-phenylethyl)-1H-indazole-3-carboxylate (2.3 g, 73 %) as a white solid. LRMS (M
-41') m/z: calcd 280.32; found 280. 1H NMR (300 MHz, d-DMS0): 6 8.08 (d, J =
8.1 Hz, 1H). 7.79 (d, ./ = 8.5 Hz, 1H), 7.43 (ddd, = 8.4, 6.9, 1.1 Hz, 1H), 7.37-7.16 (m, 6H), 6.23 (q, = 6.9 Hz, 1H), 3.93 (s, 311), 2.04-1.92 (m, 3H).
Methyl 2-(1-phenylethyl)-2H-indazole-3-carboxylate as a white solid (0.9 g).
LRMS (M-PH') m/z: calcd 280.32; found 280. 1H NMR (300MHz, c/6-DMS0): (5 7.98 (d, J= 8.1 Hz, 111), 7.84 (d, f= 8.5 Hz, 1H), 7.55-7.18 (m, 7H), 7.13-6.82 (m, 1H), 3.95 (s, 3H), 1.96 (d, ./ = 6.9 Hz, 3H).
[00214] Synthesis of 1-(1-phenylethyl)-1H-indazole-3-carboxylic acid.
CO2Me LiOH CO2H
THF/H20 , Step 3 1002151 To a mixed solution of lithium hydroxide monohydrate (0.68g, 16.8 nunol) in tetrahydrofuran (20 mL) and water (10 mL), methyl 1-(1-phenylethyl)-1H-indazole-3-carboxylate (2.3 g, 8.4 mmol) was added. The mixture was stirred at 45 C for 12 hours. Then the organic solvent was removed under reduced pressure. To the residue, 1 N
hydrochloride aqueous solution was added to adjust the pH to 3-4, and then the mixture was extracted with ethyl acetate (20 mL x 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to afford 1-(1-phenylethyl)-1H-indazole-3-carboxylic acid (1.3 g, 60 %) as a white solid, which was directly used in next step. LRMS (M+11 ) m/z: calcd 266.29; found 266.
1002161 Synthesis of ,fis/-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-1-(1-phenylethyl)-1H-indazole-3-earboxamide (Compound 112).
HO
CO2H NH2 OH HEt3N. DCM OBt, 0 r I
µ NH ,N
="' N Step 4 =µN
/
1002171 To a solution of 1-(1-phenylethyI)-1H-inda7ole-3-carboxylic acid (0.3 g, 1.12 mmol) in anhydrous dichloromethane (20 mL) was added 1H-benzo[d][1,2,31triazol-1-01 (0.14 g, 1.0 mmol), 1-ethyl-3-(3-dimethyllaminopropyl)carbodinnide hydrochloride (0.26 g, 1.37 mmol) and triethylamine (0.184 g,1.83 mmol). The mixture was stirred at room temperature for 0.5 hours, and 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (0.15g, 1.0 mmol) was added. The mixture was stirred at room temperature for 3 hours. To the reaction mixture was added water (50 mL), and the mixture was extracted with dichloromethane (20 mL X 3). The organic phase was combined, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethaneimethanol =
50:1) to give N((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-1-(1-phenylethyl)-1H-indazole-3-carboxamide (70 mg, 18%). LRMS (M+ H+) m/z: calcd. 400.47; found 400. 111 NMR (300 MHz, d6-DMS0): el 11.59 (s, 1I-I), 8.24 (s, 1H), 8.16 (d, J= 8.1 Hz, 1H), 7.68 (d, J=
8.6 Hz, I H), 7.52-7.33 (m, 1H), 7.30-7.06 (m, 6H), 6.13 (d, J= 7.0 Hz, 1H), 5.89 (s, 1H), 4.41-4.37 (m, 2H), 2.26 (s, 3H), 2.12 (s, 3H), 1.94 (d,J= 7.0 Hz, 3H).
[002181 Example 9. Synthesis of N4(2-hvdrox -4,6-di meth. Ipv ri din-3-vnincitv1)-1-(o-tolv1)-1H-indole-3-earboxamide (Compound 1131.

1.12804,91801-1 e. NaH cjir.0õ,õ.0 Me0H, rt Step 1 Br 11 SPip 2 I 0 LLIA-Br 4111P" 3 Br , I, H
Cul. '<poi NPOH THF, H20 C)0ii Ft N ncm ) I N
et,,, c step 5 Step 6 C.5./ 8 I

[002191 Synthesis of methyl 2-(2-bromophenyl) acetate.
yOH H2304, Me0H
.Br 0 S:ep _______________ 1110 Br0 [002201 A solution of o-bromophenylacetic acid (5 g, 23.2 mmol) in dry methanol (40 ml) containing concentrated sulphuric acid (I rat) was heated to reflux for 6 hours. The reaction mixture was cooled, then poured into ice water (100 mL) and extracted with ethyl acetate (30 mL
x 2). The organic layers were combined, washed successively with water (20 mL
x 2), saturated sodium bicarbonate solution (40 mL x 2) and brine. A.nd then the organic layers were combined, dried over sodium. sulfate, filtered and concentrated in vacuo to give methyl 2-(2-bromophenyl) acetate as a pale yellow liquid (5.2 g, 98%). 'H. NMR (300 MHz, CDC13): 6 7.57 (d, J = 7.8 Hz, 1H), 7.34-7.22 (m, 2H), 7.17-7.11 (m, 1H), 3.80 (s, 2H), 3.72 (s, 3H).
[002211 Synthesis of ethyl 2-(2-bromopheny1)-3-hydroxyacrylate.
OH

NaH
+ "11, H 0 Step 2 IS Br0 Br0 [002221 To a stirred solution of methyl 2-(2-bromophenyl) acetate (5.2 g, 22.7 mmol) in ethyl formate (40 mL) was added in portions the powder of sodium hydride (100%, 2.18 g, 90.8 mmol) over a period of 1 hour at 10 -15 C. After the mixture was stirred for an additional hour, the reaction was quenched with ice water (100 mL) and the two layers were separated. The aqueous layer was acidified with 10% hydrochloric acid aqueous solution and then extracted with ethyl acetate (30 mL x 3). The organic layers were combined and washed with water (2 x 20 mL), saturated sodium bicarbonate solution (2 x 40 mL) and brine in turns.
Then organic layers were dried over sodium sulfate, filtered and concentrated in vacuo to give ethyl 2-(2-bromopheny1)-3-hydroxyacrylate as a pink oil (5.33 g, 87%). Iff NMR (300 MHz, CDC13):
11.99 (d, = 12.7 Hz, 1H), 7.59 (dd, J = 7.8 Hz, 0.9 Hz, 1H), 7.46-6.98 (m, 4H), 4.25 (q, J= 7.2 Hz, 21), 1.24 (t, J = 7.2 Hz, 3H).
[002231 Synthesis of ethyl 3-(o-toluidino)-2-(2-bromophcny1)acry1ate.
e,OHOO
H
Br .4. H2N Me0H, rt N
I Step 3 Jj 1002241 To a solution of ethyl 2-(2-bromophenyI)-3-hydroxyacrylate (1 g, 3.88 mmol) in methanol (10 mL) was added o-toluidine (0.416 g, 3.88 mmol) via syringe at room temperature.
After being stirred for 18 hours at the same temperature, solvent was removed under reduced pressure to give the crude product of ethyl 3-(o-toluidino)-2-(2-bromophenyl)acrylate (1.326 g, 95%), which was used in the next step without further purification. LRMS (M
114) m/z: calcd 359.05; found 359.
1002251 Synthesis of ethyl 1-(o-toly1)-1H4ndo1e-3-carboxylate.
0 0 0.1, K3p04 is 0 DMF
N
Step 4 / 0 IP
[002261 A mixture of ethyl 3-(o-toluidino)-2-(2-bromophenyl)acrylate (1.326 g, 3.68 mmol), cuprous iodide (38 mg, 0.38 mmol), potassium phosphate (1.652 g, 7.8 mmol) and N,N-dimethylformamide (16 mL) was stirred at 75-80 C under nitrogen gas atmosphere for 16 hours.
The reaction mixture was allowed to cool to room temperature. Solvent was removed under reduced pressure. Water (16 InL) was added to the residue and the mixture was extracted with ethyl acetate (8 mL x 3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to give a residue. The residue was purified by column chromatography (silica gel, petroleum ethencthyl acctatc = 20;1) to give pure ethyl 1-(o-toly1)-1H-indole-3-carboxylate as a pale yellow solid (0.789 g, 77%). LRMS (M
+ H+) ,n/z:
calcd 279.13; found 279. 111 NMR (300 MHz, CDCI3): 6 8.26 (dd, J= 7.9, 0.7 Hz, 1H), 7.89 (d, .1= 0.7 Hz, 111), 7.53-7.11 (m, 611), 7.02 (dd, J = 8.1, 0.8 Hz, 1H), 4.43 (q, .1=7.2 Hz, 2H), 2.07 (s, 3H), 1.45 (t, J = 7.2 Hz, 3H).
1002271 Synthesis of 1-(o-tolyI)-1H-indole-3-carboxylic acid.
0 NaOH THF, H20 *0 N' OH
N Step 5 100228] A mixture of ethyl 1-(o-toly1)-1H-indole-3-carboxylate (0.789 g, 2.82 mmol), 4N
sodium hydroxide aqueous (20 mL) and tetrahydrofuran (20 mL) was stirred at 60 C for 18 hours. Most solvent was removed and the rest was acidified with 10%
hydrochloric acid aqueous to adjust to pH = 5. Pale yellow precipitate formed and was collected by filtration. The solid was washed with petroleum ether, dried in vacuo to give 1-(o-tolyI)-1H-indole-3-carboxylic acid as a pale yellow solid (0.675 g, 95%). LRMS (M + H+) m/z: calcd 251.09;
found 251.
[002291 Synthesis of N42-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-1-(o-toly1)-indole-3-carboxamide (Compound 113).
so 0 OH
EDO!, HOST OH
H2N N Et3N, DCM
N
H
Step 6 ip [002301 A mixture of 1-(o-toly1)-1H-indole-3-carboxylic acid (100 mg, 0.398 mmol), 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (60 mg, 0.398 mmol), 1-ethy1-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (114 mg, 0.597 mmol), N-hydroxybenzotriazole (81 mg, 0.597 mmol), triethylamine (81 mg, 0.796 mmol) and dichloromethane (5 mL) was stirred at room temperature for 18 hours. The solvent was removed under reduced pressure and ethyl acetate (10 rnL) was added. The resulting mixture was washed with water (10 rriL) and brine in turns and then dried over sodium sulfate.
The mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by preparative-TLC(silica gel, dichloromethane: methanol = 15:1) to give pure desired product of N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-1-(o-toly1)-1H-indole-3-carboxamide as an off white solid (100 mg, 65%). LRMS (M 10 calcd 385.18; found 385. NMR
(300 MHz, CD10D) ö 8.16 (dd, J= 6.1, 2.5 Hz, 1H), 7.87 (s, 1H), 7.44 (d, J = 4.9 Hz, 2H), 7.41-7.26 (m, 211), 7.26-7.12 (m, 2H), 6.93 (dd, J= 6.3, 2.4 Hz, 1H), 6.11 (s, 1H), 4.53 (s, 2H), 2.41 (s, 3H), 2.24 (s, 3H), 2.01 (s, 3H).
(002311 Example 10. Synthesis of compound ( or S) N-((4-me1hox -6-methv1-2-oxo-1,2-dihvdronvridin-3-v1)methvI)-2-methvl-1-(1-nhen,, 1011%, 1)-1 H-indole-3-car hoNamide (Compounds 114 and 115).
1002321 Synthesis of 3-(aminomethyl)-4-methoxy-6-methylpyridin-2-ol.

OH

\)=0 + NeCN N8HTHF

ICN ¨
UN: HCI ""=== BriEt3NCI.P0C13 Step I 0 NH2 Step 2 N.' OH Step 3 CI *NO
Me0Na, Me0H
N2H4, Raney NI
CN

_________________________________________________ &NH?
H Step 4 N OH Step 5 OH
[002331 2-amino-6-methyl-4-oxo-4H-pyran-3-carbonitrile.
NC'''CN NaH, THF
I I
Step 1 0 NH2 To a solution of malononitrile (3.3 g, 50 mmol) in anhydrous tetrahydrofuran (100 mL) was added sodium hydride (60% w/w, 2.2 g, 55 minol) at -10 C. The resultant mixture was stirred for 2 hours. Then diketene (4.2 g, 50 mmol) was added dropwise to the solution. The mixture was allowed to warm to room temperature and continued stirring for 30 minutes.
The mixture was neutralized with hydrochloric acid and then concentrated in maw to give crude 2-amino-6-methy1-4-oxo-411 -pyran-3-carbonitrile (6.0 g, 80%) as a red solid, which was used in the next step without further purification.
[00234] 2,4-dihydroxy-6-methylnicotinonitrile 10%1-1C1 CN
I
I I
0 NH2 Step 2 N OH
A suspension of 2-amino-6-methyl-4-oxo-411-pyran-3-carbonitrile (6.0 g, 40 mmol) in 10%
hydrochloride acid (60 mL) was heated under reflux for 4 hoots. The precipitate was collected by filtration and washed with water, and then recrystallized from methanol to give 2,4-dihydroxy-6-methylnicotinonitrile (5.0 g, 81%) as a brown solid. 1H NMR (300 MHz, CD30D): (512.46-12.44 (m, 1H), 11.69 (s, 111), 5.85 (s, 1H), 2.15 (s, 3H).
[00235] 4-chloro-2-hydroxy-6-methylnicotinonitrile OH CI
,1,1xCN Ax BnEt3NCI, POCI3 iCN
N OH Step 3 N OH
To a solution of 2,4-dihydroxy-6-methylnicotinonitrile (1.5 g, 10 mmol) in acetonitrile (50 mL) was added benzyltriethylammonium chloride (9.1 g, 40 mmol) and phosphorus oxychloride (6.13 g, 40 mrnol). The resulting mixture was stirred for 4 hours at room temperature. The solvent was removed by rotary evaporation. To the residue was added dichloromethane (100 mL) and water (50 mL). The organic phase was separated, washed with brine (50 mL), dried over anhydrous magnesium sulfate, filtered and concentrated to give crude product which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 4:1) to afford 4-chloro-2-hydroxy-6-methylnicotinonitrile (800 mg, 48%) as a brown solid.
1002361 2-hydroxy-4-methoxy-6-methylnicolinonitrile CI

,CN
Me0Na, Me0H
I
Step 4 To a pressure vessel was added 4-chloro-2-hydroxy-6-methylnicotinonitrile (337 mg, 2.0 mmol), sodium methoxidc (530 mg, 10.0 mmol), methanol (15 mL), and a magnetic stirrer.
The pressure vessel was sealed, and was stirred at 100 C for 16 hours before the solvent was removed by rotary evaporation. To the residue was added water (10 mL) and ethyl acetate (50 mL). The organic layer was separated and concentrated in vacuo to provide crude product which was purified by column chromatography (silica gel, dichloromethane/methanol = 40:1) to afford 2-hydroxy-4-methoxy-6-methylnicotinonitrile (70 mg, 21%) as a brown solid.
1002371 3-(aminomethy1)-4-methoxy-6-rnethylpyridin-2-01 N2H4, Raney Ni CN
I Et0H, 60 C NH2 N OH Step 5 = N OH

2-Hydroxy-4-methoxy-6-methylnicotinonitrile (70 mg, 0.43 mmol) was dissolved in ethanol (10 mL) and warmed to 60 C before it was treated with raney nickel (0.5 mL slurry in water) followed by addition of hydrazine monohydrate (2 mL). The resultant mixture was allowed to stir at 60 C for 2 hours. The cooled reaction mixture was filtered through celite and rinsed with methanol. The filtrate was concentrated in vacuo to provide crude product which was purified by column chromatography (silica gel, dichloromethane/methanol = 10:1) to afford (aminomethyl)-4-methoxy-6-methylpyridin-2-ol (40 mg, 56%) as a white solid.
LRMS (M
m/z: calcd 168.09; found 168. HPLC purity (214 nm): 73%. IH NMR (300 MHz, d5-DMS0): 6 6.04 (s, 1H), 3.77 (s, 3H), 3.42 (s, 2H), 2.15 (s, 3H).
[082381 Synthesis of N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-2-methyl-1-(1-phenylethy1)-1H-indole-3-carboxamide (Compound 138).

(!) \ H2N NH HOBT, EDCI, Et3N, DCM, rt, HN-=

To a solution of 2-methyl-1-(1-phenylethyl)-1H-indole-3-carboxylic acid (66 mg, 0.24 mmol) in anhydrous dichloromethane (5 mL) was added N-hydroxybenzotriazole (38 mg, 0.29 mmol), 1-ethyl-343-(dimethylamino)propylicarbodiimide (55 mg, 0.29 mmol) and trimethylarnine (36 mg, 0.36 mmol), and stirred at room temperature for 0.5 hour, 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(1H)-one (40 mg, 0.24 mmol) was added and stirred at room temperature for 16 hours. To the reaction mixture was added water (10 mL), extracted with dichloromethane (10 mL) 2 times, combined and concentrated the organic layers, the residue was purified by column chromatography (silica gel, dichloromethane/methanol = 20:1) to give N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1 -phenylethyl)-1H-indole-3-carboxami de as an off-white solid (0.08 g, 52%). LRMS (M H+) m/z: calcd 429.21; found 429.
1002391 Synthesis of (R or S) N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-1H-indole-3-earboxamide (Compounds 114 and 115).

o o o HO _ _ Fir*..\ 0 IN..../.....\HN
/ HN.,. \ A / 0 Olt ChiraisHtepPLO5 separatiorii 0 .....( r i ,. 0 _...- N
I
ss. ' (R or S) I (S or R) . it N
N46-hydroxy-2-methoxy-4-methylpyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-1H-indole-3-carboxamide (80 mg, 0.19 tnmol) was separated by chiral HPLC (LA(AD50)-TEA20rnin, then (R or S) N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-1H-indole-3-carboxamide (25 mg, 31%) and (S or R) N-((4-m.ethoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)m.ethyl)-2-methyl-1-(1-phenyiethyl)-1H-indole-3-carboxamide (15 mg, 19%) were obtained. The retention, times were 8.222 minute ("Peak 1";
Compound 114) and 13.531 ("Peak. 2"; Compound 115) minute respectively in chiral prep-HPLC
chromatography.
LRMS (M 4-.H.1) m/z: calcd 429.21, found 429. HPLC Purity (214 nm): 100%. 1H
NMR (300 MHz, d6-DMS0): 6 11.59 (s, 1H), 7.75-7.72 (m, 2H), 7.35-7.25 (m, 3H), 7.15 (d,./= 7.5 Hz, 2H), 7.10-7.07 (m, 1H), 7.02-6.93 (in, 2H), 6.15 (s, 1H), 5.96 (q, J= 6.9 Hz, 1H), 4.33 (d, J' 5.1 Hz, 2H), 3.85 (s, 31-I), 2.62 (s, 3H), 2.20 (s, 3H), 1.89 (d, j = 7.5 Hz, 3H).
,Exatnnle 11., Synthesis of compound N-((4.6-dimeth v1-2-oxo-1,2-dify,dropyridin-3-vInnethvI)-2.6-dimethyl- 7-oxo-1-( I -phenviciliv1)-6,7-didivdro-.1 11 -nvrrolol 2.3-0 pyridine-3-carboxamide (Compound 139).

. H
ci, MelNaH/DIvt% r y O e/Nti Cl/Et0H I r- .,. )t.....õ..ko NaHITHF N
...... 0 2 F
-"" No, Stop) -1-"*. NO2 Step2 1 Ster.s3 0 CI 6; o 0 (7-3, iii r ,.44-.. i Step4 \ _____ co,co3romF ...,, --11--1 LiowEtow 420 ¨ o 11 "-- - µi it4....
.
¨ND I.. or- step'. - VX.0 ---N H
4 0 N=" d HATESEt3N _ --H

ster38 04 , N
1002401 Synthesis of 4-chloro-l-methyl-3-nitropyridin-2(1.11)-one H I
Cr.õ10 MeliNalliDMF N 0 _ (TX
NO2 5teP1 NO2 CI a To a stirred solution of 4-chloro-3-nitropyridin-2(1H)-one (3.0 g, 17 mmol) in N,N-dimethylformamide (50 ml,) was added sodium hydride (60% wlw, 1.0 g, 25.5 mmol) in batches at 0 C. The mixture was stirred at room temperature for 30 minutes. Then iodomethane (2.9 g, 20.4 mmol) was added dropwise to the above solution at room temperature .The resultant solution was stirred at room temperature for 12 hours. Once starting material was consumed, the reaction mixture was quenched with ice water (100 mL) at 0-10 C, and extracted with ethyl acetate(100 mL x 3).The organic phase was washed with brine (100 mi., x 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethanelmethanol = 10:1) to give 4-chloro- 1-methy1-3- nitropyridin-2(1H)-one (3 g, 94%) as a yellow solid. LRMS (M + fl+) m/z: calcd 188.0; found 188.
[002411 Synthesis of ethyl 2-(1.-methy1-3-nitro-2-oxo-1,2-dihydropyridin-4-y1)-oxobutanoate N. 00 )c)(0-- NaHITHF NO
'NO2 Step2 CI
To a stirred solution of ethyl 3-oxobutanoate (2.5 g, 19 mmol) in tetrahydrofuran (50 mL) was added sodium hydride (60% w/w, 0.96 g, 23.9 mmol) in batches at 0 C. The mixture was stirred at room temperature for 30 minutes. Then the solution of 4-chloro-l-methy1-3-nitropyridin-2(1H)-one (3.0 g, 16 mmol) in tetrahydrofuran (50 mi,) was added in one portion. The resultant solution was stirred and heated to 50 C for 12 hours. Once the starting material had been consumed, the reaction solution was quenched with water (100 mL) at 0 C, and extracted with ethyl acetate (100 mI, x 3). The combined organic layers were washed with brine (100 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/m.ethanol =
10:1) to give ethyl 2-(1.-methyl-3-nitro-2-oxo-1,2-dihydropyridin-4-y1)-3-oxobutanoate (2.5 g, 56%) as a yellow solid. LRMS (M H+) ,n/z: calcd 282.09; found 282.
[002421 Synthesis of ethyl 2,6-dimethy1-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-clpyridine-3-carboxylate NO2 Fe/NH4CtiEtatH20 \ r-0 Step3 To a solution of ethyl 2-(1-methy1-3-nitro-2-oxo-1,2-dihydropyridin-4-y1)-3-oxobutanoate (2.5 g, 8.8 mmoi) in ethanol (50 mL) was added ammonium chloride (0.5 g, 9 rnmol) in water (5 mL) at room temperature .The mixture was stirred and heated to reflux. Then iron powder (0.5 g, 8.9 mmol) was added in one portion. The mixture was stirred at reflux for 2 hours.
Once starting material was consumed, the resultant mixture was filtered when it was hot, and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 10:1) to give ethyl 2,6-dimethy1-7-oxo-6,7-dihydro-1H-pyrrolo [2,3-c]pyridine-3-carboxylate (1.5 g, 75%) as a brown solid. LitMS (M + 1.14) m/z: calcd 234.1;
found 234. NMR (400 MHz, c/6-DMS0): 6 12.54 (s, 1H), 7.30 (d, = 5.1 Hz, 1H), 6.78 (d, J
= 5.1 Hz, 1H), 4.24 (q, J = 5.1 Hz, 2H), 3.51 (s, 3H), 2.56 (s, 3H), 1.32 (t, J= 5.1 Hz, 3H).
[00243] Synthesis of ethyl 2,6-dimethy1-7-oxo-1-(1-phenylethyl)-6,7-dihydro-IH-pyrrolo12,3-c] pyridine-3-carboxylate (.1 H
Sr Cs2CO3/DRIF
¨N 8 .................... - 0 Step4 To a stirred solution of ethyl 2,6-dimethy1-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridine-3-carboxylate (1.5 g, 6.4 mmol) in N,N-dimethylformamide (50 mL) was added (1-bromoethyl)-benzene (1.5 g, 7.9 rrunol) and cesium. carbonate (3.14 g, 9.6 mmol). The resultant solution was stirred at 80 C for 12 hours. Once starting material was consumed, the reaction mixture was quenched with water (50 mL), and extracted with ethyl acetate (100 mL x 3).
The combined organic layers were washed with brine (100 mi., * 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 5:1) to give ethyl 2,6-dimethy1-7-oxo-141-phenylethyl)- 6,7-dihydro-1H-pyrrolo[2,3-c]pyridine-3-carboxylate (1.5 g, 69%) as a yellow solid. LRMS (M + H.) m/z: calcd 338.16; found 338.
[00244] Synthesis of 2,6-dimethy1-7-oxo-1-(1.-phenylethyl)-6,7-dihydro-1H-pyrrolo[2,.3-clpyridine- 3-carboxylic acid *
0 ms.r...., LiOHIEt0H/H20 0 N
_Nb¨irr st,p5 ¨Nct\ H

A mixture of ethyl 2,6-di meth y1-7-oxo- l -(1-phenylethyl)-6,7-dihydro-1H-pyrrolo [2,3-c]pyridine-3-carboxylate (500 mg, 1.5 mmol) and lithium hydroxide (50 mg, 2.1 mmol) in ethanol (20 mL) and water (5 mL) was stirred and heated at 80 C for 12 hours.
Once starting material was consumed, the mixture was concentrated and the residue was dissolved into water (50 mL). Then the solution was extracted with ethyl acetate (20 mL x 2).The aqueous layer was collected, adjusted to pH 4, and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with brine (20 mL x 2), dried over anhydrous sodium sulfate, filtered and concentrated to give 2,6-dimethyl- 7-oxo-1-(1-phenylethyl)-6,7-dihydro-1H-pyrrolo[2,3-c]pyridine-3-carboxylic acid (200 mg, 44%) as a light yellow solid which was used directly in the next step without further purification. LRMS (M + Fr) miz: calcd 310.13;
found 310.
[00245] Synthesis of N-((4,6-dimethy1-2-oxo-1,2-di hydropyridin-3-yl)methyl)-2,6-dimethy1-7-oxo- 1-(1-phenylethyl)-6,7-dihydro-1H-pyrroloi 2,3-cl pyridine-3-carboxamide (Compound 139).
#11 p Th .,_-_ 0 31 HATU/Et3N , 0 N , H2N\,.. .s., I
Step6 0 mH
\ _ \...___... z.rx N \ /
o o To a mixture of 2,6-dimethy1-7-oxo-1-(1-phenylethyl)-6,7-dihydro-1H-pyrrolo [2,3-c]pyridine-3-carboxylic acid (200 mg, 0.64 mmol) and 4-(aminomethyl)-3,6-dirnethylpyridin-2(111)-one (118 mg, 0.78 mmol) in N,N-dimethylfonnamide (20 mL) was added o-(7-azabenzotriazol-1-y1)-N,N,Isr,N-tetramethyluronium hexafluorophosphate (365 mg, 0.96 mmol) and triethylamine (388 mg, 3.84 mmol) at room temperature. The resultant mixture was stirred at room temperature for 2 hours. Once starting material left was consumed, the reaction mixture was quenched with water (50 mL), and extracted with ethyl acetate (50 mL * 3).The combined organic layers were washed with brine (100 mL * 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 1:2) to give N((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)- 2,6-dimethy1-7-oxo-1-(1-phenylethyl)-6,7-di hydro-1H-pyrrolo [2,3-c] pyridine-3-carboxamide (150 mg, 53%) as a white solid. LRMS (NI + m/z: calcd 444.22; found 444. HPLC
purity (214 nm): 97%.1H NMR (400 MHz, CD3OD): ö 7.22-7.13 (m, 4H), 7.01 (d, J= 7.2 Hz, 2H) , 6.69 (d, J= 7.2 Hz, 110, 5.99 (s, 1H), 4.35 (s, 21), 3.50-3.21 (m, 41), 2.26 (s, 3H), 2.13 (s, 3H), 2.06 (s, 3H), 1.82 (d, J= 7.2 Hz, 3H).
[00246] Example 12. Synthesis of compound (R)-N4(2-hydroxv-4,6-dimethvipyridin-1:1)methvI)-6-methvl-2-(methvlamino)-7-( I -phinviethyl)-7H-pvrrolo12,3-dip-vrimidine-5-carboxamide (Compound 140).
j'step D4PEA, EtOti P4(0A02, LiCIACO. DMF = m ,,, Step 2 .... -C.e 0 õ,õ
N., EDCI.H08t. EtyN NH
= Step 4 Niekõ.11 A )--=N " 2 sne-75-4.
y,,.1 )--N=
="*.N = OH 0 N
[00247] Synthesis of (R)-5-bromo-2-chloro-N-(1-phenylethyl)pyrimidin-4-amine Br:(NH2 fR) ()PEA. Et0H __ Ci)cr els1H
I.
CI' '14 'CI So Step 1 To a solution of 5-bromo-2,4-dichloropyrimidine (5 g, 22 mmol) and (R)-I-phenylethanamine (2.7 g, 22 mmol) in ethanol (50 mL) was added N,N-diisopropylethylamine (4.3 g, 33 mmol).
The reaction solution was stirred at room temperature for 12 hours. The resultant mixture was concentrated in vacuo. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl. acetate = 6:1) to give (R)-5-bromo-2-chloro-N-(1-phenylethyppyrimidin-4-amine as a white solid (5 g, 74%). LRMS (M + Er) m/z: calcd 310.98; found 310. NMR
(300 MHz, CDC13): ô 8.10 (s, 1H), 7.41-7.25 (m, 5H), 5.73 (d, J = 6.9 Hz, IH), 5.39-5.34 (m, 1H), 1.61 (d, J= 6.6 Hz, 3H).
[00248] Synthesis of (R)-methyl 2-ehloro-6-methyl-7-(1-phenylethyl)-711-pyrrolo[2,3-d I pyrimidin e- 5-ca rboxylate N../..\1Br 0 N \
02, UCI, K2CO3, DR,IF
CI Pdt0A
N 67NH *
Step 2 CI
I
/
A. solution of (R)-5-brom.o-2-chloro-N-(1-phenylethyl)pyrimidin-4-amine (5 g, 16 mm.o1), methyl but-2-ynoate (3.1 g, 32 mmol), lithium chloride (690 mg, 16 mmol), potassium carbonate (5.5 g, 40 nunol.) and palladium acetate (360 mg, 1.6 mm.ol) in N,N-dimethylformamide (50 mL) was degassed and back-filled with nitrogen for three times, then heated at 120 C
for 4 hours. The reaction mixture was filtered and the filtrate was concentrated in maw, extracted with ethyl acetate (50 mL), washed with water (50 mL), dried over anhydrous magnesium sulfate and purified by column chromatography (silica gel, petroleum ether/ethyl acetate =
5:1) to give (R)-methyl. 2-chloro-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylate as a yellow oil (800 mg, 15%). LRMS (M Hi) m/z: calcd 329.09; found 329. 1H. NMR
(300 MHz, CDC13): (5 9.16 (s, 1H), 7.39-7.18 (m, 5H), 6.45-6.42 (m, 1. fl), 3.96 (s, 3H), 2.57 (s, 3H), 2.05 (d, = 7.2 Hz, 311).
1002491 Synthesis of (R)-2-chloro-6-niethy1-7-(1-phenylethyl)-711-pyrrolo[2,3-dipyrimidine-5- carboxylic acid TcMeat THF
N N
(R) Step 3 .R) = =õ

Lithium hydroxide anhydrate (882 mg, 21 mmol) in water (3 mL) was added to (R)-methyl 2-chloro-6-methy1-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylate (700 mg, 2.1 mmol) in tetrahydrofuran (5 mL) and methanol (10 mL) and the resultant mixture was stirred at room. temperature for 12 hours. The mixture was evaporated, added with water (5 mL), acidified with aqueous hydrochloric acid (1M) to pH = 2. The precipitate solid was filtered and dried to obtain (R)-2-chloro-6-methy1-7-(1-phenylethyl)-711-pyrrolo[2,3-d] pyrimidine-5-carboxylic acid as a white solid (500 mg, 75%). LRMS (M H4) m/z: calcd 315.08; found 315.
[002501 Synthesis of (R)-2-chloro-N-((2-hydroxy-4,6-ditnethylpyridin-3-y1)methy1)-6-methyl-7- henylethyl)-7H-pyrrolo [2,3-d I pyrimidine-5-carboxamide N =-=, ED(.71.HOSI, Et3N rW) + a t4H2 _________ N Step 4 N N /
OH
OH 0 s'N
/
To a solution of (R)-2-chloro-6-methy1-7-(1-phcnylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylic acid (100 mg , 0.32 mmol) in dichloromethane (10 mL) was added with hydroxybenzotriazole (65 mg, 0.48 mmol), 1-Ethy1-3-(3-dimethylaminopropypethyl-carbodiimide hydrochloride (92 mg, 0.48 mmol) and triethylamine (97 mg, 0.96 mmol). After stirred for 30 minutes, 3-(aminomethyl)-4,6- dimethylpyridin-2-ol (49 mg, 0.32 mmol) was added and the reaction mixture was stirred at room temperature for 12 hours.
The solution was concentrated, diluted with water (20 mL), extracted with ethyl acetate (20 mL). The organic layers were separated, combined, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 20: 1) to afford (R)-2-chloro-N-((2- hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-mcthyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidinc-5-carboxamide (100 mg, 70%). LRMS (M + 11+) nilz: calcd 449.16; found 449.
1002511 Synthesis of (R)-N4(2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-6-methyl-(methylamino)- 7-(1-pheny1ethy1)-7H-pyrrolo12,3-dlpyrimidine-5-earboxamide (Compound 140).
/ tki Step 5 N N N
OH (3 --t4 OH 0 H
A solution of (R)-2-chloro-N-((2-hydroxy-4,6-dimethylpyridin-3-ypmethyl)-6-methyl-7-(1-phenyl ethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (30 mg , 0.07 mmol) in methylamine (1 M in tetrahydrofuran, 3 mL) was stirred at 150 C for 30 minutes under microwave (pressure:
17.2 bar, equipment power: 150W). The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethane/methanol = 10: 1) to afford (R)-N-((2-hydroxy-4,6- dimethylpyridin- 3-yl)methyl)-6-methyl-2-(methylamino)-7-(1-phenylethyl)-7H-pyrrolo [2,3-d] pyrimidine-5-carboxamide (10 mg, 34 %). LRMS (M + 171-) calcd 444.23;
found 444. HPLC purity (214 mu): 92%. 11.1 NMR. (300 MHz, CD30D): 6 8.55 (s, 1H), 7.30-7.23 (m, 5H), 6.15-6.10 (m, 2H), 4.47 (s, 2H), 2.90 (s, 3H), 2.40 (s, 3H), 2.38 (s, 3H), 2.24 (s, 3H), 2.03 (d, J= 7.2 Hz, 3H).
1002521 Example 13. Synthesis of co mound (R)-N4(2-hydroxy.-4,6-diniellivinyridin-3-y1)methyl)-2-methoxy-6-nieihvi-7-(1-nhea v1ethyb-7H:pyrrolot2,3-dipyri in i ne-carboxamide (Compound 141).

HyT'' \--1)NaOMe, KileOH
_______________________________________ * H
N -IN\ I\ Step 1 A solution of (R)-2-chloro-N4(2-hydroxy-4,6-dimethylpyridin-3-ypmethyl)-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (70 mg, 0.15 mmol) in Me0H (1 mL) solution of sodium methanolate (20 mg) was stirred at 100 C for 60 minutes under microwave (pressure: 15.3 bar, equipment power: 150W). The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethane/methanol = 10 :
1) to afford (R)-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methoxy-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (37 mg, 51.9%). LRMS (M + H.1") miz:
calcd 445.21; found 445. HPLC purity (214 urn): 100%. 1H NMR (400 MHz, CD30D) 6 8.75 (s, 1H), 7.29 ¨ 7.22 (m, 5H), 6.11 ¨ 6.07 (m, 2H), 4.46 (s, 2H), 3.90 (s, 3H), 2.48 (s, 3H), 2.36 (s, 3H), 2.21 (s, 3H), 2.05 (d, J= 7.2 Hz, 3H).
1002531 Example 14. Synthesis of (R)-N4(2-hydroxv-4.6-di inethvinvridin-3-vDmettivi)-6-me th v1-741-phenviethyl)-2-(ovrrolid n-l-v1)-7H-oyrrolo12.3-dlovri Ifni di ne-5-earboxamid e (Compound 142).
CNH
f'7 _______________________________________ 41' N N N Step 1 N N N
CI
OH 0 A solution of (R)-2-chloro-N42-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl- 741-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (70 mg, 0.15 mmol) in pyrrolidine (1.0 inL) was stirred at 150 C for 30 minutes under microwave (pressure: 12.2 bar, equipment power: 150W). The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethane/methanol = 10: 1) to afford (10-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-7-(1-phenylethyl)-2-(pyrrolidin-l-y1)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (45 mg, 60.0%). LRMS (M + Fr) m/z: calcd 484.26; found 484. HPLC
purity (214 nm): 97.8%. NMR
(400 MHz, CD3013) 6 8.56 (s, 11), 7.27 - 7.20 (m, 5H), 6.06 - 6.02 (m, 2H), 4.45 (s, 2H), 3.52 - 3.45 (m, 4H), 2.40 (s, 3H), 2.35 (s, 3H), 2.21 (s, 3H), 2.01 - 1.94 (m, 711).
[002541 Example 15. Synthesis of compound (R)-N-((2-hydroxy-4,6-dimethylpyridin-3-vbmethyl)-6-methvI-2-morph oil n 0-741-ph e nylethvI)-714-pyrrolo12.3-d 1pyrimidine-5-earboxamide (Compound 143).
µ-.2/ 0r--\NH
N' ''''sirsY*- H i)-N N N Step 1 N

L/.0 A solution of (R)-2-chloro-N4(2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-6-methyl-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (70 mg, 0.15 mmol) in morpholine (1.0 mL) was stirred at 150 C for 30 minutes under microwave (pressure: 10.5 bar, equipment power: 150W). The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethane/methanol = 10: 1) to afford (R)-N-02-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-2-rno rpho I i no-7-(1-phenylethyl)-7H-pyrrolo [2,3-d]pyrimi dine-5-carboxamide (55 mg, 70.5 %). LRMS (M miz:
calcd 500.25; found 500. HPLC purity (214 nm): 98.6%. 11-1 NMR (400 MHz, CD301)) 6 8.65 (s, 1H), 7.28 (dt, .1= 11.9, 7.7 Hz, 5H), 6.10 (dd, .1= 13.7, 6.5 Hz, 2H), 4.51 (s, 211), 3.78 - 3.65 (m, 811), 2.47 (s, 311), 2.41 (s, 311), 2.26 (s, 311), 2.03 (d, 1= 7.2 Hz, 3H).
[002551 Example 16.
Synthesis of compound (R)-2-amino-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-741-phenylethyl)-711-pyrrolo12,3-dlpyrimidine-5-carboxamide (Compound 144).

N N N N N N
OH 0 OH 0 --N))--NH2 A solution of (R)-2-chloro-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-7- (1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (70 mg, 0.15 mmol) in ammonium hydroxide (5 mL) was stirred at 150 C for 30 minutes under microwave (pressure: 17.2 bar, equipment power : 150W). The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethanelmethanol = 10: 1) to afford (R)-2-amino-N4(2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (25 mg, 38 %). LRMS (M + Hi) m/z: calcd 430.21;
found 430.
HPLC purity (214 nm): 93%. 1H NMR (300 MHz, CD30D): 6 8.57 (s, 1H), 7.34-7.19 (m, 5H), 6.19-6.17 (m, 1H), 6.10 (s, I H), 4.47 (s, 2H), 2.33 (s, 3H), 2.30 (s, 3H), 2.23 (s, 3H), 1.99 (d, J=
7.2 Hz, 3H).
1002561 Example 17. Synthesis of N-(0.6-4 imetlz v1-2-oxo-I th-opv rid in-3-vl)mettry1)-2-methyl-1 -(phenvIsulfon.0-1 H-indole-3-cat. Mixa ( COMPO tt 45).
HOBt EDCI >:N OH 0 Et:N 11,0 HO\ \>--, NH N DC:5M 4- NH / NH +
I

0 0 so NaH /NI , THF
Jo i00257] Synthesis of N-((2-hydroxy-4,6-dimethylpyridin-3-yOmethy1)-2-methy1-1H-indole-3-carboxamide HOBt MCI
Et3N
1-10)T. DCM =
NOH H
_N
step 3 0 ()Dr 10 To a solution of 2-methyl- I FI-indole-3-carboxylic acid (3.3g, 18.85 mmol), I -hydroxybenzotriozole (4.69g, 37.7 mmol, 2eq), 1-(3-dimethylaminopropyI)-3-ethykarbodiimide hydrochloride (7.24 g, 37.7 mmol, 2eq), triethylamine (20.0 mL) in dichloromethane (100 mL) was added 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (5.77g, 37.7 mmol, 2eq).
The reaction mixture was stirred at 20 C for 13 hours. The mixture was washed with water (20 mL x 2). The organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to give the residue, which was purified by column chromatography (silica gel, dichloromethane/methanol = 10:2) to give N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide as a white solid (3.00 g, 9.7mmo1, 51%). LRMS
(M + H ,n/z:
ealed 309.36; found 310. HPLC Purity (214 nm): >95%. 1H NMR (300 MHz, d6-DMS0):
11.61 (s, 1H), 11.43(s, 1H) 7.74 (d, J = 5.1 Hz, 1H), 7.58-7.54 (m, 1H), 7.31 (d, = 7.2Hz, 1H), 7.04 (t, J = 6Hz, 3H), 5.89 (s, 1H), 4.32 (d, J= 4.5 Hz, 2H), 2.57 (s, 3H), 2.26 (s, 3H), 2.12 (s, 3H).
[00258] Synthesis of N-((4,6-dimethy1-2-oxo-1,2-dihyd ropyridin-3-yi)methyl)-2-methyl-1-(phenylsulfonyl)-1H-indole-3-carboxamide (Compound 145) 11.0 NaH HN
/
¨ 9,0 NH
THF N,s, To a solution of N-((4õ6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide (0.15g, 0.485mmo1) in tetrahydrofuran(10 mL) was added sodium hydride (0.023 g, 0.582 mmol) at 0 C. The mixture was stirred at 0 C for 30 minutes. Then benzenesulfonyl chloride (0.086 g, 0.485 mmol) was added to the mixture. The reaction was warmed to room temperature slowly and stirred for another 30 minutes. It was quenched by adding water (10 mL) slowly, and extracted by ethyl acetate (10 mL x 3). The organic layer was dried and purified by Prcp-HPLC to afford N -((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-mcthyl-1-(phenylsulfony1)-1H-indole-3-carboxamide (9 mg 4.1% yield). LRMS (M + H+) m/z:
calcd 449.14; found 450. HPLC Purity (215 nm): 97%.1H NMR (400 MHz, CDC13): 6 8.25 (d, J =
6.8Hz, 1H), 7.81 (dd, J = 8.4 Hz, J = 0.8 Hz, 2H), 7.62-7.7.54 (m, 2H), 7.46-7.40 (m, 2H), 7.35-7.28 (m, 2H), 6.80 (t, J = 6Hz, 1H), 6.51 (s, 1H), 4.58 (d, J = 4.6 Hz, 2H), 2.80 (s, 3H), 2.64 (s, 3H), 2.45 (s, 3H).
1002591 Example 18. Synthesis of compound N-(12-4droxy,-4,6-dimethvinvridin-3-yl)methvl)-2-methv1-1-(1-o henvlethvI)-1H-pvrrolot2,3-blpyridine-3-c ar bo N.
a In i e (COMDOUnd 1461.
o o Ct BINOL, 50 C
:-ONle .....................
Cs2CO3, ONISO cx7... +
)0. Br jaH.
NH
0. \ io Step i Step 2 HOBT
1.10H, THF/Me0H/H20 Et. CH, N \
__________ = I 4. s=-= NH? _________________ f Step 3 N c OH Step 4 N OH
1002601 Synthesis of methyl 2-methyl1.H-pyrrolo[2,3-b] pyridine-3-carboxylate Cul, BINOL, 50 C OMe I
I Cs2CO3, DMS0 OMe _________________________________________ , N NH2 Step 1 Nr N
To a solution of 3-iodopyridin-2-amine (5.5 g, 25 mmol.) in dimethylsulfoxide (40 mL) were added methyl acetoacetate (3.48 g, 30 mmol), copper iodide (476 mg, 2.5 mmol), 1,1'-binaphthy1-2,2'-diol (1.43 g, 5.0 mmol) and cesium carbonate (8.15 g, 25 mrn.o1). The resultant mixture was stirred at 50 C for 4 hours. To the reaction mixture was added ethyl acetate (400 mL). The organic phase was washed with brine (80 mL x 3), dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to give crude product which was purified by column chromatography (petroleum ether/ethyl acetate = 4:1) to afford methyl 2-methyl-IFI-pyrrolo[2,3-b]pyridine-3-carboxylate (1.02 g, 21.4%) as a white solid. IF.1 NMR (300 MHz, CD3OD): ô 8.46-8.43 (m, 1H), 7.61-6.69 (m, 211), 3.89 (s, 31-1.), 2.73 (s, 3H).
1002611 Synthesis of methyl 2-methyl-1-(1 -phenylethyl)-1H-pyrrolo [2,3-b I
pyri dine-3-carboxylate 0, 0 ¨OW Br -OMe NaH, DMF
+
Step 2 N N
To a cooled (0 C) solution of methyl 2-methyl-1H-pyrrolo[2,3-bjpyridine-3-carboxylate (1.02 g, 5.36 mmol) in N,N-dimethylformamide (20 mL) was added sodium hydride (60% w/w, 236 mg, 5.90 mmol). The resultant mixture was stirred for 15 minutes. Then (1-bromoethyl)benzene (2.00 g, 10.8 mmol) was added and the reaction was allowed to warm to room temperature. The reaction was maintained at ambient temperature for 12 hours. The reaction mixture was poured into saturated ammonium chloride solution (100 mL) with stirring. The mixture was extracted with ethyl acetate (200 mL x 2) and the combined organic phase was washed with brine, dried over magnesium sulfate, filtered, and concentrated to give crude product which was purified by column chromatography (petroleum ether/ethyl acetate = 20:1) to afford methyl 2-methy1-1-(1-phenylethyl)-1H-pyrrolo[2,3-13]pyridine-3-carboxylate (500 mg, 31.7%) as a viscous oil. 11-1 NMR (300 MHz, CD30D): 68.38-8.35 (m, I H), 8.25-8.23 (m, I H), 7.30-7.14 (m, 6H), 6.55-6.48 (m, 1H), 3.88 (s, 3H), 2.54 (s, 3H), 2.02 (d, J= 7.2 Hz, 3H).
1002621 Synthesis of 2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-Npyridine-3-carboxylic acid.

OH
I \Li0H, THF/MeOHM20 N N = Step 3 N
To a solution of methyl 2-methyl-1-(1-phenylethyl)-1H-pyffolo[2,3-b]pyridine-3-carboxylate (500 mg, 1.70 mmol) in tetrahydrofuran (10 mL), methanol (20 mL) and water (4 mL) was added lithium hydroxide (163 mg, 6.80 mmol). The resultant reaction mixture was stirred at 60T for 48 hours. The mixture was concentrated in WIC110. Then the residue was diluted with water (40 mL) and slowly acidified with IN hydrogen chloride to pH = 4-5. The mixture was extracted with ethyl acetate (100 int, x 3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated to give 2-methy1-1-(1-phenylethyl)-1H-pyrrolo[2,3-b]ppidine-3-carboxylic acid as a white solid (400 mg, 84.0%).
1002631 Synthesis of N4(2-hydroxy-4,6-dim ethylpyridin-3-yl)methy1)-2-methyl-1-(1-phenylethyl)-1H- pyrrolo[2,3-b]pyridine-3-carboxamide (Compound 146).

OH EDCI, Et3N, 9 NH2 ________________________________________ 1 N N) (t7. N OH Step 4 µ.
To a solution of 2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-b]pyridinc-3-carboxylic acid (400 mg, 1.43 mmol) in dichloromethane (30 mL) were added 1-hydroxybenz,otriazole (291 mg, 2.15 mmol), 1-(3-dimethylaminopropyI)-3-ethylcarbodiimide hydrochloride (412 mg, 2.15 mmol) and triethylamine (434 mg, 4.30 mmol). The resultant mixture was stirred at room temperature for 30 minutes. Then 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (262 mg, 1.72 mmol) was added and the resultant mixture was stirred at room temperature for 16 hours. Water (50 ml) was added to the mixture. The mixture was extracted with dichloromethane (100 rnL x 2). The organic layer was concentrated in vacuo to provide crude product which was purified by column chromatography (silica gel, dichloromethanel methanol = 20:1) to afford N4(2-hydroxy-4,6-di methy 1pyri din-3-yl)methyl)-2-methyl-1-(1-phe ny lethyl)-1H-pyrrolo- [2,3-b]pyridi ne-3-carboxamide (400 mg, 67.6%) as a white solid. LRIvIS (M H.) m/z: calcd 414.21;
found 414.
HPLC purity (214 nm): 94%. IHE NMR (300 MHz, CD3OD): 6 8.22-8.13 (in, 2H), 7.28-7.15 (m, 6H), 6.47-6.45 (m, 1H), 6.09 (s, 1H), 4.50 (s, 2H), 2.42 (s, 3H), 2.39 (s, 3H), 2.23 (s, 3H), 2.02 (d, ..1= 7.2 Hz, 3H).
1002641 Example 19. Synthesis of compound l-benzvl-N4(2-hydroxv-4.6-dimethvloviidin-3-vbmethyl)-2-methyl-1H-indole-3-ca rho x a mide (Compound 119).
meo2c lio2c CO2Me Br Cs2CO3 NaOH
DAW THFikle0H/H20 Step 1 Step 2 / t4 HOBT. MCI 01-1 0 .õ.0H
Et3N, DCM HN
=
Ni-12 \
Step 3 N
\ I
[00265] Synthesis of methyl 2-methyl-1H-indole-3-carboxylate Me02C
CO2 Me / Br Cs2CO3 DMF
+
N Step I-N

To a solution of methyl 2-methyl-1H-indole-3-carboxylate (378 mg, 2 mmol) in N,N-dimethylformamide (10 mL) was added (bromomethAbenzene (340 mg, 2 mmol) and cesium carbonate (652 mg, 2 mmol), then stirred at 60 6C for 12 hours. The reaction mixture was concentrated in vacuo. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 1:1) to give methyl 2-methyl-I H-indole-3-carboxylate (446 mg, 80%). LRMS (M + H') ni/z: calcd 279.13; found 279.
1002661 Synthesis of 1-benzy1-2-methy1-1H4ndole-3-carboxylic acid Me02C HO2C
NaOH
THF/Me01-111-120 N *
Step 2 N
\
To a solution of methyl 1-benzy1-2-methyl-1H-indole-3-carboxylate (446 mg, 1.6 mmol) in tetrahydrofuran (20 mL) and methanol (7 mL) was added sodium hydroxide (320 mg, 8 mmol) in water (7 mL), then stirred at 60 6C for 12 hours. The reaction mixture was concentrated in vacuo and acidified to PH = 4 with 6 N hydrochloric acid, collected and dried to give 1-benzy1-2-methy1-1H-indole-3-carboxylic acid as a white solid (212 mg, 50 %). LRMS (M +
m/z: calcd 265.11; found 265.
1002671 Synthesis of 1-benzyl-N-((2-hydroxy-4,6-di methylpyridin-3-yl)methy1)-2-methyl-1H-i ndole- 3-carboxamide (Compound 119).

4. N OH HOB?, EDO OH 0 NH?
Et3N, DCM N
¨N 411 N
To a solution of 1-benzy1-2-methyl-1H-indole-3-carboxylic acid (212 mg, 0.8 mmol) in dichloromethane (20 mL) was added 1H-benzo[d][1,2,3]triazol-1.-ol (135 mg, 1 m.mol), 1-(3-Dimethylaminopropy1)-3-ethylcarbodiimide hydrochloride (190 mg, I mmol) and triethylamine (252 mg, 2.5 mmol), and stirred at room temperature for 0.5 hour, 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (152 mg, I mmol) was added and stirred for 4 hours. To the reaction mixture was added water (20 mL), which was extracted with dichloromethane (2 x 20 mL), combined and the organic layers were concentrated, the residue was purified by column chromatography (dichlorom.ethane/methanol = 20:1) to afford 1-benzyl-N-((2-hydroxy- 4,6-dimethylpyridin-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide (200 mg, 63%).
LRMS (M +
Fr) m/z: calcd 399.19; found 399. HPLC purity (214 nm): 100%. 111 NMR (300 MHz, d6-DMS0): 6 11.61 (s, 1H), 7.80-7.69 (in, 211), 7.46-7.42 (m, 111), 7.30-7.22 (m, 3H), 7.11-7.07 (m, 2H), 6.98 (d, f = 7.5 Hz, 2H), 5.89 (s, 111), 5.46 (s, 2H), 4.33 (d, J= 4.5 Hz, 2H), 2.57 (s, 3H), 2.26 (s, 3H), 2.11 (s, 3H).
002681 Example 20. SI, nillesis of compound N-((2-hydroxv-4,6-dimethylpyridin-vi)methvi)-2-methvl- 1 1-pbenylethl, 1)- 1 i-l-pµ rrolof 3,2-blvyridine-3-carboN amide (Compound l26.
Me02C
Br LION
0 0 i,Cu20, Cs2C0s. 110"C Me:OH/1120 UNI12+ )ciLe 40 Step 1 Step 2 1102c:
HATO N¨ I-IN + N0H
Et3N, DMF
I mi2 _______ N Step 3 N
100269) Synthesis of methyl 2-m ethyl-141 -phenylethyl)-1H-pyrrolo13,2-b ipy ridine-3-ca rboxylate Mf:02C
N I 0 0 NH2 Br i,Cu20, Cs2CO3, DMF, 110 C
)LA0". +
N
Step 1 To a solution of 2-iodopyridin-3-amine (500 mg, 2.27 mmol), cuprous oxide (32 mg, 0.23 mmol), cesium carbonate (740 mg, 2.27 mmol) in N,N-dimethylformamide (100 mL) was added methyl methacrylate (290 mg, 2.5 mmol). The reaction solution was stirred at 110 C for 12 hours. Then the reaction mixture was cooled with an ice bath and sodium hydride (60% in oil, 91 mg, 2.27 mmol) was added under. The resulting mixture was stirred at room temperature for half an hour. Then (1-bromoethyl)benzene (418 mg, 2.27 mmol) was added. Then the mixture was stirred at room temperature for 1 hour. After the reaction was completed, it was quenched with water (100 mi.), and extracted with ethyl acetate (50 mi,x 3). The combined organic phase were washed with water (20 rnLx 3) and brine (20 mL), dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (silica gel, petroleum ether/ethyl acetate =
6:1) to give methyl 2-methyl- 1-(1-phenylethyl)-1H-pyrrolo[3,2-b]pyridine-3-carboxylate (80 mg, 12%). LRMS (M + m/z: calcd 295.14; found 295.

1002701 Synthesis of 2-inethyl-1-(1-phenylethyl)-1H-pyrrolo[3,2-blpyridine-3-carboxylic acid Me02C HO2C
LiOH
Me0H/H20 N * Step 2 N
To a solution of methyl 2-methy1-1-(1-phenylethyl)-1H-pyrrolo[3,2-14yridine-3-carboxylate (80 mg, 0.27 mmol) in methanol (3 mt.) and water (1 mL) was added lithium hydroxide (57 mg, 1.36 mmol). The reaction mixture was stirred with refluxing for 15 hours. The mixture was adjusted pH 3 with IN aqueous hydrochloric acid. The aqueous phase was extracted with dichloromethane (50 mL x 3). The organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue, which was purified by column chromatography (silica gel, dichloromethane/medianol = 20:1) to give 2-methyl-1-(1-phenylethyl)-1H-pyrrolo- [3,2-b]pyridine-3-carboxylic acid (20 mg, 26%). LRMS
(M if) m/z:
calcd 279.12; found 279.
1002711 Synthesis of N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-2-methyl-1-(1-phenylethyl)-1H- pyrrolo[3,2-blpyridine-3-carboxam id e (Compound 126) HO2Ci HATO
=-,Tr.N,y0H H
Et3N, DItAF, /
N-tt, Nr0 T Stgp 3 N-- N-s=
The mixture of 2-methyl-I -(1-phenylethyt)-1H-pyrrolo[3,2-b]pyridine-3-carboxylic acid (20 mg, 0.11 mmol), 2-(1H-7-Azabenzotriazol-1-y1)--1,1,3,3-tetramethyl uronium hexafluorophosphate Methanaminiurn (50 mg, 0.13 mmol), 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (19 mg, 0.12 mmol) and triethylamine (23 mg, 0.22 mmol) in N,N-dimethylformamide (5 mL) was stirred for 12 hours. After the reaction was completed, the reaction was diluted with ethyl acetate (100 mL), and washed with water (20 mLx 3) and brine (20 mL), dried over anhydrous sodium sulfate and concentrated to give the residue, which was purified by column chromatography (silica gel, dichloromethaneJmethanol = 30:1) to give N4(2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl- I -( I -phenylethyl)-1H-pyrrolo[3,2-b]pyridine-3-carboxamide (15 mg, 33 %).
LRMS(M-FH ) m/z: calcd 415.20; found 415. HPLC Purity (214 nm): 99%. 1H NMR
(300 MHz, d6-DMS0) 611.46 (s, 1H), 9.49 (t, ii = 4.2 Hz, J2=8.1 Hz, 1H), 8.28 (d, J= 3.6 Hz, 1H), 7.50 (d, = 6.3 Hz, 1H), 7.36-7.27 (m, 3H), 7.16 (d, J = 5.7 Hz, 1H), 7.03-7.00 (m, 1H), 6.03 (m, 1H), 5.85 (s, 1H), 4.37 (s, 2H), 2.89 (s, 3H), 2.50 (s, 3H), 2.10 (s, 3H), 1.89 (d, J= 5.4 Hz, 3H).
100272] Example 21. Synthesis of (R or S)-1-sec-butvl-N42-hydroxy-4,6-di methvbwridin-3-0 )methyl)-2-methyl-M-indole-3-carbox a mide (Compound 147) and (S
tir Th.. I -sec-butO-N-((2-hyd roxy-456-di Elfiethylpyridin-3-11 nethi,1)-2-met it v. iisr-in carboNaniiile (Compound 148).
mec,c;
CO2Me LiOH
THF/Me0H/H2 0 4 Cs2CO3, DMF
==-= tq Step I __ 10/ -Step 2 NCh HOST. EDO OH 0 Et1N. DCM
tip NH2 __ Step 3 Chrial HPLC
Step 4 * N
1002731 Synthesis of methyl 1-sec-butyl-2-methyl-M-indole-3-carboxylate M
p02Me e02C
Cs2CO3, DMF
Step 1 To a solution of methyl 2-methyl-/H-indole-3-carboxylate (0.5 g, 2.6 mmol) in N,N-dimethylformamide (20 mL) was added cesium carbonate (1.7 g, 5.2 mmol) and 2-bromobutane (0.71 g, 5.2 rnmol), the mixture was stirred at 100 C under microwave for 1 hour, the mixture was concentrated and purified by column chromatography (silica gel, ethyl acetate/petroleum ether = 1:20) to give methyl 1-sec-butyl-2-methyl-1H-indole--carboxylate (141 mg, 22%).
[002741 Synthesis of 1-sec-butyl-2-methyl-/H-indole-3-carboxylic acid Me02C iOH HO-C
L
MeOHIH20 - ¨
step 2 -N
To a mixed solution of methanol (10 mL) and water (2 mL), methyl 1-sec-butyl-2-methyl-M-indole-3-carboxylate (141 mg, 0.58 mmol) and lithium hydroxide (100 mg, 2.4 mmol) were added. The mixture was stirred at room temperature for 12 hours. Then the reaction mixture was acidified by hydrochloric (1 A1) to adjust pH = 6 and extracted with dichloromethane (10 mL x 3). The organic layers were combined and concentrated to give 1-sec-buty1-2-methyl-/H-indole-3-carboxylic acid (97 mg, 72%).
1002751 Synthesis of 1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-methyl-M-indole-3-carboxamide (Compound 123) 1402c N HO N¨ HN
ip+ H2 _____ Step 3 A mixture of 1-sec-butyl-2-methyl-Hi-indole-3-carboxylic acid (97 mg, 0.42 mmol), 1-ethy1-3-(3¨climethyllaminopropyl)carbodiimide hydrochloride (121 mg, 0.63 mmol), N-hydrox3rbenzotrizole (85 mg, 0.63 mmol) and triethylamine (127.26 mg, 1.26 mmol) in dichloromethane (30 mL) was stirred for 30 minutes at room temperature. Then to the mixture, 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (63.8 mg, 0.42 mmol) was added. The resulting mixture was stirred at room temperature for 12 hours. Then the mixture was washed with water (20 mL x 3). The organic layer was concentrated to give a residue which was purified by column chromatography (silica gel, dichloromethane/methanol = 20:1) to give 1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide (57 mg, 30%).
LR.MS (M m/z: calcd 365.2; found 365. HPLC purity (214 nm): 99%. 1H.NMR
(300 MHz, CD30D): ö 7.72-7.69 (m, 1H), 7.53 (d, J = 7.2 Hz, 1H), 7.11-7.06 (m, 2H), 6.10 (s, 1H), 4.53-4.51 (m, 3H), 2.62 (s, 3H), 2.41 (s, 311), 2.24 (s, 3H), 1.95-1.93 (m, 2H), 1.59 (d, J = 6.9 Hz, 3H), 0.72 (t, J= 7.5 Hz, 3H).
1.902761 Synthesis of (R or S)-1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-2-methyl-/H-indole-3-carboxamide and (S or R)-1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-y1) methyl) -2-methyl-iii-indole-3-carboxamide / Chrial HPLC
__________________________ =
\ ) \ ) R or S S or R
1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-/H-indole-carboxamide (35 mg, 0.1 mmol) was separated by chiral. prep-HPLC (Daicel IA
(200 mm x 20 mm x 5 um), hexane: ethanol (0.2% DEA) = 70: 30, flow rate: 19 mLimin), then (R or S) 1-sec-butyl-N4(2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-/H-indole-3-carboxamide (11 mg, 30%) and (S or R)-1-sec-butyl-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-M-indole-3-carboxam ide (6 mg, 16%) was obtained. The retention times were 8.030 minutes ("Peak 1"; Compound 147) and 14.126 minutes ("Peak 2"; Compound 148) respectively in chiral HPLC chromatography. LRMS (M H') tri/z: calcd 365.2; found 365. HPLC purity (214 nm):
99%. IH.NMR (300 MHz, CD30D): ö 7.73-7.70 (m, 1H), 7.56-7.53 (m, 1H), 7.12-7.09 (m, 2H), 6.11 (s, 1H), 4.54 (s, 3H), 2.64 (s, 3H), 2.43 (s, 3H), 2.25 (s, 3H), 1.97-1.90 (m, 2H), 1.61 (d, J =
6.9 Hz, 3H), 0.73 (t, J = 7.5 Hz, 3H).
[002771 Example 22. Synthesis of compound N42-hydroxv-4,6-dimethylpyridin-3-v1)metliv1)-2-mettiv1-1-phenvl- 1H-indole-3-carboxamide (Compound 133).
HO.
8 Cu(04.02, 4A MS NaOH
HN 1+ ________________________ ' (D-N __________ (X-/ N
0 DMAP, E't3N ¨ 0 C2H5OH/H20 OH
0 step 1 0 step 2 0 HOBT, EDCI
+ ("IA, N ____________ 411.' N H
DCM,tt3N N N
step 3 0 OH
[002781 Synthesis of methyl 2-methyl-1-phenyl-1H-indole-3-earboxylate HO OH
Y"--2 Cu(0A02, 4A MS __ HN
+ _______________________________________ b- 1/ ----N
( I DMAP, Et3N ----/
0 step.' o A mixture of methyl 2-methyl-1H-indole-3-carboxylate (500 mg, 2.65 mmol), phenylboronic acid (384 mg, 3.17mmo1), diacetylcopper (453 mg, 3.98 mmol), triethylamine (0.44 ml, 3.98 mm.o1), N,N-dimethy1pyridin-4-amine (486 ml, 3.98 mmol) and 4A molecular sieve (1.02 g) in dichloromethane (15 ml.,) was stirred at room temperature for 12 hours. After filtration, the mixture was concentrated and purified by chromatography (silica gel, petroleum: ethyl acetate =
10:1) to afford methyl 2-methyl-I -phenyl- IH-indole-3-carboxylate as white solid (272 mg, 39%).11-1NMR (400 MHz, CDC13) 6 8.19 (d, J= 8.0 Hz, 1H), 7.66 ¨ 7.51 (m, 3H), 7.36 (dd, J =

5.3, 3.2 Hz, 2H), 7.30 (s, 1H), 7.21 ¨7.14 (m, 1H), 7.04 (d, .1= 8.2 Hz, 1H), 4.00 (s, 3H), 2.62 (s, 3H).
[002791 Synthesis of 2-methyl-1-phenyl-1H-indole-3-carboxylic acid NaOH

r NI( 2 5 2 W H
0 step 2 0 To a solution of methyl 2-oxo- 1 -pheny1-1,2-dihydropyridine-4-carboxylate (272 mg, 1.03 mmol) in alcohol/ water (v/v = 1/1, 5 mL) was added lithium hydroxide (62 ing, 1.55 mmol), and the mixture was stirred at room temperature for 8 hours. The reaction mixture was concentrated, acidified by diluted aqueous hydrochloric acid (1 N, 5 mL) and extracted with ethyl acetate (2 x 10 ml). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 2-methyl- 1-phenyl- I H-indole-3-carboxylic acid as white solid (115 mg, 44%). LRMS (M-H+) miz: calcd for 251.09, found 251.
[002801 Synthesis of N4(2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-2-methyl-1-phenyl-M-indole-3-carboxamide (Compound 133).

HOBT, EDC1 OH I DCM,EtaN N
step 3 0 OH
To a solution of 2-methyl-1 -pheny1-1H-indole-3-carboxylic acid (115 mg, 0.46 mmol) in anhydrous dichloromethane (3 mL) was added 1H-benzo[d][1,2,3]triazol-1-ol (75 mg, 0.55 mmol), 1-Ethy1-3-(3-dimethyllaminopropyl)carbodiimide hydrochloride (106 mg, 0.55 mmol) and triethylamine (0.16 mL, 1.15 mmol). The mixture was stirred at room temperature for half an hour, 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (70 mg, 0.46 mmol) was added and stirred at room temperature for 3 hours. The reaction mixture was added water (20 mL), extracted with dichloromethane (20 mL x 2), combined and concentrated the organic layers to give the residue, which was purified by column chromatography (silica gel, dichloromethane/methanol = 25:1) to afford N-((2-hydroxy-4,6-dimethylpyridin-3-Amethyl)-2-methy 1-1 -pheny1-1H-indole-3-carboxamide (12 mg, 50% yield. LRMS (M-41') m/z: calcd for 385.18, found 385.

(300 MHz, CDC13)43 12.62 (s, 1H), 7.89 (d, J= 8.0 Hz, 1H), 7.77 ¨7.40 (m, 3H), 7.43 ¨ 7.18 (m, 3H), 7.22 ¨7.00 (m, 2H), 5.98 (s, 1H), 4.65 (s, 2H), 2.60 ¨ 2.52 (m, 3H), 2.45 (s, 3H), 2.25 (s, 3H).
1002811 Example 23. Synthesis of compound N-(0,6-dianettiv1-2-oxo-1,2-ditivilropyridiii-3-vDmethili-2-inethyl-1-(pvrimidin-5-vhnethyl)-1 H-ind o le-3-ea. r box amide (Compound ,N14 DCM moo r/ Ce2CO3, D1AF
N N
Q.141-3 ""-Q't*I' Step 1 0 Step 2 0 N
N
N=f KOH 011 HORT. EDCE
MeOH/H20 Et3N, DCM rig _____________ HO HN I N
Step 3 Step 4 N /

1 0 0 2 8 2 1 Synthesis of 5-(bromoniethyl)pyrimidine PBr3, DCM B r Step I
To a solution of pyrimidin-5-ylmethanol (0.5 g, 4.5 mmol) in dichloromethane (50 m.1.,) was added phosphorus tribromide (0.6 g, 2.25 nunol) at 0 C. The solution was stirred at room temperature for 12 hours. The solution was washed with sodium bicarbonate and concentrated to give a residue, which was purified by column chromatography (silica gel, ethyl acetate/petroleum ether = 1:10) to give 5-(bromomethyl)pyrimidine (0.3 g, 39%).
[002831 Synthesis of methyl 2-methyl-1.4pyrimidin-5-ylmethyl)-111-indole-3-earboxy1ate NH
N Br Cs2CO3, DMF
Me0 Me0 0 Step 2 147-=-/
To a solution of methyl 2-methyl- /H-indole-3-carboxylate (0.5 g, 2.6 mmot) in N,N-dimethylformarnide (20 mL) was added cesium carbonate (1.7g, 5.2 mmol.) and 5-(brornomethyl)pyrimidine (0.3 g, 1.7 minol). The mixture was stirred at 100 C
for 1 hour and then concentrated to give a residue. The residue was purified by column chromatography (silica gel, ethyl acetate/petroleum ether = 1:20) to give methyl 2-methyl-1-(pyrimidin-5-ylmethyl)-111-indole-3-carboxylate (110 mg, 14%) 1002841 Synthesis of 2-methyl-1-(pyrimidin-5-ylmethyl)-/H-indole-3-carboxylic acid KOH
Me0 Me0H/H20 õ
N Step 3 HO

Nr-4 To a mixed solution of methanol (10 mL) and water (2 mL), methyl methyl 2-methyl-1-(pyrimi din-5-ylmethyl)-/H-indole-3-carboxylate (110 mg, 0.39 mmol) and potassium hydroxide (50 mg, 0.98 mmol) was added. The mixture was stirred at reflux for 12 hours. Then the reaction mixture was acidified by hydrochloric acid aqueous solution (1N) to adjust pH 6 and extracted with dichloromethane (10 nciL * 3). The organic layers were combined and concentrated to give 1-isopropy1-2-methyl-/H-indole-3-carboxylic acid (60 mg, 58%).
[002851 Synthesis of N-04,6-dimethy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-2-methyl-1-(pyrimidin- 5-ylmethyl)-/H-indole-3-carboxamide (Compound 149) 411# OH HOST, EDGE
Et3N, HO N DCM NH2 H I PI 1 Step 4 N

A mixture of 2-methyl-1-(pyrimidin-5-ylmethyl)-M-indole-3-carboxylic acid (60 mg, 0.22 mmol), 1- ethyl-3-(3-dimethyllaminopropyl)carbodiimide hydrochloride (65 mg, 0.34 mmol), N-hydroxybenzotrizole (180 mg, 1.33 mmol) and triethylamine (46 mg, 0.34 mmol) in dichloromethane (30 mL) was stirred for 30 minutes at room temperature. Then to the mixture, 3-(aminomethyl)-4,6-dimethyl pyridin-2-ol (33.44 mg, 0.22 mmol) was added. The resultant mixture was stirred at room temperature for 12 hours. Then the mixture was washed with water (20 mL x 3). The organic layer was concentrated to give a residue which was purified by column chromatography (silica gel, dichloromethane/methanol = 20:1) to give N4(4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl-1-(pyrimidin-5-y1methy1)411-indole-3-carboxamide (17 mg, 19%). LRMS M + calcd 401.19; found 401. HPLC purity (214 nm):
92%.
'H.NMR (300 MHz, CD30D): ö 9.03 (s, 1H), 8.43 (s,2H),7.80-7.78 (m, 1H), 7.39-7.36 (m, 1H), 7.19-7.16 (m, 2H), 5.56 (s, 2H), 4.55 (s, 2H), 2.62 (s, 3H), 2.42 (s, 3H), 2.45 (s, 3H).

Example 24. Synthesis of compound 1-benzoyi-N4(2-hydroxv-4,6-diniethvipyridin-vt nnet 1)-2-methyl- l H-indole-3-carboxamitit (Compound y_o NH Naii THF 0)r. / 1110 Ci stepl HOSt Et3N :"14 OH
DCM N Ns, OH DCM /
N N
step2 NH2 step3 S
[002861 Synthesis of tert-butyl 1-benzoy1-2-methyl-1H-indole-3-carboxylate \it 91, o / NH + NaH
)-Lsci THF \/-0 N
0 ij stepl so 0 IP
To a suspension of sodium. hydride (73 mg, w/w = 60%, 1.8 mrnol) tert-butyl 2-methy1-1H-indole- 3-carboxylate (350 mg, 1.5 mmol) in tetrahydrofuran (30 mL) was added benzoyl chloride (250 mg, 1.8 mmol). The reaction solution was stirred at room temperature for 12 hours.
The mixture was concentrated to give a residue, which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 3:1) to give tert-butyl 1.-benzoy1-2-m.ethyl.-1H-indole-3-carboxylate (440 mg, 88%).
[002871 Synthesis of 1-benzoy1-2-methyl-111-indole-3-ca rboxylic acid 'LoNO

TFA
DCM
, HO\ N\ µL./
0 step2 .A mixture of tert-butyl 1-benzoy1-2-methy1-1H-indole-3-carboxylate (440 mg, 1.3 mmol), lithium hydroxide monohydrate (276 mg, 6.6 mm.o1), tetrahydrefuran (12 mL), methanol (4 mL) and water (4 mL) was stirred at room temperature for 4 hours. The mixture was concentrated, acidified to pH = 2 with concentrated hydrochloric acid and extracted with ethyl acetate (20 mL
x 3). The organic phase was dried over anhydrous sodium sulfate, filtered and concentrated to give 1-benzoy1-2-methy1-1H-indole-3-carboxylic acid (260 mg, 72%). LRMS (M + 1-14) m/z:
calcd 279.13; found 279.
1002881 Synthesis of 1-benzoyl-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methy1)-methyl-1H-indole- 3-carboxamide (Compound 150) HOER
0 EDC __NI 0H 0 N
m NNy0H gn, H 110 H * N
4. I steP3 To a solution of 1-benzoy1-2-methy1-1H-indolc-3-carboxylic acid (125 mg, 0.45 mrnol), 1-hydroxybenzotriozole (122 mg, 0.9 mmol), 1-(3-dimethylarninopropy1)-3-ethylcarbodiimide hydrochloride (173 mg, 0.9 mmol), triethylamine (0.4 mL) in dichloromethane (20 mL) was added 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (137 mg, 0.9 mmol). The reaction mixture was stirred at 20 C for 13 hours. The mixture was washed with water (20 mL x 2).
The organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to give the residue, which was purified by column chromatography (silica gel, dichloromethanelmethanol 10:1) to give 1-benzoyl-N-((2-hydroxy-4,6-di methylpyridi n-3-yl)methyl)-2-methyl-114-i ndo le-3-carboxamide as a white solid (50 mg, 53%). LRMS (M + H4) m/z: calcd 413.17;
found 413.
HPLC Purity (214 rim): 96%. 111 NMR (300 MHz, d6-DMS0): 11.57 (s, 111), 8.13 (q, J = 5.1 Hz,1H), 7.74-7.70 (m, 411), 7.60-7.58 (m, 2H), 7.20-7.00 (m, 3H), 5.88 (s, 1H), 4.32 (d, J = 5.1 Hz, 211), 2.37 (s, 3H), 2.267 (s, 3H), 2.12 (s, 3H).
[00289) Examnle 25, Synthesis of eomnound N-((-1,o-diniethµ 1-2-oxo-1,2-dilivdronvridin-3-vOmethvb-2-methvi-1(1-oxo-1-(Dvrrolidin-1-0)propan-2-v1)-- 1 H dole-3-carboxamide (Compound 151).
0 0 H Cs2CO3 DCM DMF
&ep ___________________ ,Tkr"\ + Me * __________ Step 2 0 1--J 0 OMe NaOH 0 * OH
HOST, EDCI
ON
THF/Me0H/1120 C Et3N, DCM }¨k= N-Ar.N , 0 N, '=== NH2 Step 3 Step 4 .*
OH
\
0 0 ¨
(002901 Synthesis of 2-bromo-1-(pyrrolidin-l-yl)propan-1 -one DCM Y
ytsci 4. (NO
Step 1 Br Br Pyrrolidine (0.12 mol, 8.3 g) was added to stirred solution of 2-bromopropanoyl chloride (0.58 mol, 10 g) in dichloromethane (200 ml) at 0 C. After 0.5 hour, the reaction mixture was warmed to room temperature, stirred for 2 hours, and saturated ammonium chloride solution (20 ml) was added. The solvent was evaporated and the mixture was extracted with ethyl acetate (200 ml).
The organic phase was separated, dried over anhydrous sodium. sulfate, filtered and concentrated to dryness. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 55:1) to give 2-bromo-1-(pyrrolidin-l-yl)propan-l-one (9.8 g, 93%). LCMS M -4- H4) ni/z: calcd 205.01; found 205.
[002911 Synthesis of methyl 2-methyl-1.-(1.-oxo4-(pyrrolidin-1-yl)propan-2-y1)-1H-indole -3-carboxylate Cs2CO3 DMF 0 410.
Me0 \ ____________ r CN 11\r N
Step 2 Br OMe To a solution of methyl 2-methyl-1H-indole-3-carboxylate(0.52 mmol, 0.1 g) in NN-dimethylformamide (5 inL), 2-bromo-1-(pyrrolidin-l-yppropan-1-one (0.52 mmol, 0.137 mg) and cesium carbonate (1.05 mmol, 384 mg) was added. The reaction mixture was heated at 100 C for 12 hours. LC-MS showed the start material was consumed. The solvent was evaporated and the residue was washed with water (10 ml), extracted with dichl.orom.ethane (20 m1). The organic layer was separated, concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethanelmethanol = 45:1) to give methyl 2-methyl-1-(1-oxo-1-(pyrrolidin-l-yl)propan-2-y1)-1H-indole-3-carboxylate (100 mg, 93%). 1H NMR (300 MHz, CD30D): 6 8.07-8.03 (m., if!), 7.44-7.40 (m, 1H), 7.19-7.14 (m, 2H), 5.53-5.49 (m, 1H), 3.90 (s, 3H), 2.80 (s, 3H), 2.00-1.80 (m, 2H), 1.74 (d, J = 5.1 Hz, 3H), 1.67-1.64 (m, 4H), 1.32-1.28 (m, 2H).
[002921 Synthesis of 2-methy1-1-(1-oxo-1-(pyrrolidin-1-y1)propan-2-y1)-1H-indole-3-carboxylic acid 0 NAOH 0 __ THFIMe0H/H20 <./
Step 3 To a solution of sodium hydroxide solution (50 mg, 1.2 mmol) in tetrahydrofuran, menthol and water (20 mL, 3:1:1, V/V) was added methyl 2-methy1-1-(1-oxo-1-(pyrrolidin-1-y1)propan-2-y1)-1H-indole-3-carboxylate (100 mg, 0.318 mmol). The reaction mixture was stirred at 70 C for 10 hours. The mixture was quench with 10% hydrochloric acid aqueous (2 mL), extracted with dichloromethane and menthol (60 mL, 10:1). The combine organic layer was dried by anhydrous sodium sulfate, filtered and concentrated to give 2-methyl-1-(1-oxo-1-(pyrrolidin-1-Apropan-2-y1)-1H-indole-3-carboxylic acid (90 mg, 92%). LCMS (M ) nilz: calcd 300.15;
found 300.
190293] Synthesis of N-04,6-dimethy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-2-methyl-1-(1-oxo-1- (pyrrolidin-1-yl)propan-2-y1)-1H-indole-3-carboxamide (Compound 151).
OH ,0N
0 HOBT, EDCI
N, Cl1/4.1-1(r.N N Et3DCM 0 ___________________________ NH2 N
, I
Step 4 HN I i OH

2-Methyl-1-(1-oxo-1-(pyrrolidin-l-y1)propan-2-y1)-1H-indole-3-carboxylic acid (90 mg, 0.30 mmol) was dissolved in dichloromethane (15 mL), and then N-hydroxybenzotrizole (0.45 mmol, 60 mg), 1-(3-dimethylaminopropy1)-3-ethylcarbodiimide hydrochloride (0.45 mmol, 86 mg), and triethylamine (1.69 mmol, 2 ml) was added to the mixture. After the mixture was stirred at room temperature for 10 minutes, 3-(aminomethyl)-4, 6-dimethylpyridin-2-ol (0.30 mol, 50 mg) was added. The mixture was stirred at room temperature for 18 hours. Then washed with. water (20 nit), extracted with dichloromethane (20 mL). The organic layer was separated, and concentrated to give a residue and the residue was purified by column chromatography (silica gel, dichloromethane/methanol = 15:1) to give N4(4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yOmethyl)-2-methyl-1-(1-oxo-1-(pyrrolidin-1-y1)propan-2-y1)-1H-indole-3-carboxami de (60 mg, 70%). LCMS (M H.1) m/z: calcd 434.23; found 434. HPLC Purity (214 am): 99%.

(300 MHz, DMSO-d6) 8 11.62 (s, 1H), 7.75-7.72 (m, 2H), 7.42 (d, J 5.7 Hz, 1H), 7.10-7.06 (m, 2H), 5.89 (s, 1H), 5.49-5.44 (m, 1H), 4.32-4.30 (m, 2H), 3.42-3.32 (m, 3H), 3.28-3.16 (m, 1H), 2.61 (s, 3H), 2.26 (s, 3H), 2.12 (m, 3H), 2.02-1.96 (m, 1H), 1.70-1.57 (m, 3H), 1.42(d, J =
5.1 Hz, 3H).
[00294] Example 26. Synthesis of Compounds 326, 327, 346 and Related Compounds and Intermediates. The title compounds of this Example and other related compounds were prepared according to the following general scheme. In addition, where indicated, modifications of this scheme are disclosed for the synthesis of still additional related compounds of the invention and intermediates thereof.

(S)-2-rnethylpropane-2-suifinamide
9 -;.T.S'NH2 0, p-N
""\ 0 Ti(OEt)4 MehlgElr . : \\_._/¨\ p lo--NC.)..._ktf. Y___ H
/ 60-90% 16 A tr \.,...7--= 70-86% _ 01¨
Step 1 04- d>9;1 hiN-8:
' Step 2 0 Step 4 X Cy \,....,0 .? I .1 = -I
1.) HCI, 95% 6 O\ HH Br 2.) NahIC03 HOAc. Et0H, 70%.
Step 3 NH2 Step 5 ry1/2%-(R or S) (R or S) >ro RuPhos Precat ittr_CN- NaOH
--40 \ 1Sr¨CN4 Gen III 0 N
N
Step 7 I
Step 6 ../olif.: HO

(R or S) (R or S) ..,...%,:tlo Ht!1 1 NH2 \r"014 Nt./___CNH
R19 _., 0 N HCI

.. HN ii OMe, Me, Et, OCF2 1 . _,.r 1 Step 9 * 0 Step 8 6 o (R or 5) (1 or S) Step : .CWit20 N

- HN i ril, 1 411 (R20 is H or an optional N-substituen0 (R or S) Step 1: (S,E)-tert-butyl 4-(((tert-butylsulfinAimino)methyl)piperidine-1-carboxylate:
(S)-2-methylpropane-2-isirlfinamide II
)s, r-õ. N ii2 Ot 0 Ti(OEt).$
H
0--(-- 95% 0-+

To a round bottomed flask charged with a magnetic stir bar was added (S)-2-methylpropane-2-sulfinamide (20,46 g, 169 mmol), tert-butyl 4-formylpiperidine-1-carboxylate (30 g, 141 mmol), DCM (300 mL), and Ti(0E04 (59.0 ml, 281 mmol). The solution was stirred at room temperature for 3 h before it was quenched with brine (80 mL). The solution was stirred for 30 minutes before filtering. The filter cake was washed with DCM and the filtrate was placed in a separatory funnel and washed with water. The organics layer was dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue solidified to the title compound (29 g, 92 mmol, 65.1 % yield) raiz 217.
[002951 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 1 using the appropriate starting materials and modifications.
Name Structure ink (S,E)-2-methyl-N-((tetrahydro-2H-pyran-4- S¨N

yl)methylene)propane-2-sulfinamide 0, (S,E)-2-methyl-N-((tetrahydro-2H-thiopyran-4- 'S¨N

yl)methylene)propane-2-sulfinamide H
( )-(E)-2-methyl-N-((3-methyloxetan-3- 001--4, 204 vi jrnethylene)proparte-2-sulfinamide N--S
1002901 Step 2: Tert-butyl 4-((S)-14(R or S)-1,1-dimethylethylsolfinamido)ethyl) piperidine-l-earboxylate:
%S¨N MeMgE3r 4-0 H 65- 85%
0 dr> 9:1 HN¨S
To a round bottomed flask charged with a magnetic stir bar was added (S,E)-tert-butyl 4-((tert-butylsulfinylimino)methyl)piperidine-i -carboxylate (36.4 g, 115 minol), DCM
(400 mL), and the solution was cooled to 0 C in an ice bath with stirring. To this solution was added MeMgBr (77 nil, 230 mmol) (3M in diethyl ether) and the reaction stirred for 4 h while warming to room temperature. The reaction was carefully quenched via the addition of saturated aqueous The solid were broken up by the addition of IN Ha The layers were separated and the aqueous phase was extracted with DCM. The combined organics phase was dried over Na2SO4, filtered, and concentrated in VaC140 to afford the title compound (29 g, >9:1 dr) which is used without further purification in the next step.
[002971 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 2 using the appropriate starting materials and modifications.
Name Structure miz (S)-2-methyl-N-((R or S)- I -(tetrahydro-2 El-pyran-4- (0-4 V¨

= 234 yl)ethyl)propane-2-sulfinamide HN-S
'r) (S)-2-methyl-N-((R or S)-1-(tetrahydro-2H-thiopyran-4- 80¨?

yl)ethyl)propane-2-sulfinamide ( )-2-methyl-N-(1-(3-methyloxetan-3-ypethyl)propane-2-sulfinamide 220 µ0 1002981 Step 3: (R or S)- tert-butyl 4-(1-aminoethyl)piperidine-l-earboxylate:
HC
-->"
e-NO¨e Y-HN-1, ..2 To a 1 L round bottomed flask charged with a magnetic stir bar was added crude tert-butyl 4-((S)-1-((S)-1 ,1-dimethylethylsulfinamido)ethyppiperidine-1-carboxylate (29 g) was taken up in Me0H (200 mL) before addition of a 4 N solution of RCA in 1,4-dioxane (24.06 ml, 96 mmol).
The resulting solution was then stirred at room. temperature for 1 h at rt.
The methanol was then removed in vacuo to afford viscous oil which was treated with sat'd aqueous NalIC03 (¨ 500 mL) and extracted with ethyl acetate (2 x 500 mL). This organic phase was combined, dried with MgSO4, filtered, and solvent was then removed in vacuo affording the title compound (22 g) which was used without further purification.
1002991 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 3 using the appropriate starting materials.
Name Structure nifz Ds.?
(R or S)-1-(tetrahydro-2H-pyran-4-ypethanamine 130 (R or S)-1-(tetrahydro-2H-thiopyran-4-yl)ethanamine S 146 coqle ( )-1-(3-methyloxetan-3-yl)ethanamine 116 [003001 Step 4: Methyl 2-(2-bromopheny1)-3-oxobutanoate:
Li 8r ur iisast 0 .0" 0 A round bottomed flask was charged with a magnetic stir bar and methyl 2-(2-bromophenyl)acetate (25 g, 109 mmol) and THF (50 mL). This solution was cooled to -78 C
before drop wise addition of a 1M solution of LiHMDS in THF (218 ml, 218 mmol). The reaction was stirred for 30 min at -78 C before addition of 1-(1H-imidazol-1-yl)ethanone (14.42 g, 131 mmol) dissolved in a mixture of THF:DMF (112 mL THF, 24 mL DMF). The solution was stirred for 1 h before quenching with sat'd aqueous NH4C1 (-250 mL) and diluting with Et0Ac. The layers were separated and the aqueous phase was extracted with Et0Ac (-2 x 250 mL). The combined organic extract was washed with brine, dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography using an eluent of ethyl acetate/hexanes (10:1) to afford methyl 2-(2-bromopheny1)-3-oxobutanoate (32.5 g, 102 mmol, 93 % yield).
[003011 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 4 using the appropriate starting materials.
Name Structure m/z Br 0 methyl 2-(2-bromo-4-chloropheny1)-3-oxobutanoate ________________________________________________ 110 = 304 ci Or methyl 2-(2-bromo-4-methoxypheny4-3-oxobutanoate methyl 2-(2-bromo-4-fluoropheny1)-3-oxobutanoate 0 289 ( [003021 Step 5: (R or S, Z)-teri-butyl 4-(1-0-(2-brontopheny1)-4-tnethoxy-4-oxobut-2-en-2-ylarnino)ethyl)piperidine-1-carhoxylate:

Br 0 -*.'0 (110 \ 0 HI I
Its 0-N34 HOAc. Et0H, 70%

y'D-45' >1-0 To a round bottomed flask was added (R or S)-tert-butyl 4-(1-aminoethyl)piperidine- 1-carboxylate (9.35 g, 40.9 mmol), Et0H (75 mL), and methyl 2-(2-bromopheny1)-3-oxobutanoate (7.40 g, 27.3 mmol) (from Step 4). To this solution was added AcOH (1.563 ml 27.3 mmol) and the reaction was heated overnight at 85 C before cooling to room temperature and concentrating. The crude residue was purified via silica gel chromatography (330g, 100%
hexanes to 25% EA in hexanes) to afford the title compound (6.45 g, 13.40 mmol, 49.1 % yield).
1003031 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 5 using the appropriate starting materials.
Name Structure m/z Ni (R or S,Z)-methyl 2-(2-bromopheny1)-34(1-((l 1 .
u I 383 HN
(tetrahydro-2H-pyran-4-yl)ethyl)amino)but-2-enoate &Iv' i a (R or S,Z)-methyl 2-(2-bromo-4-chloropheny1)-3-(( I - I Br 41 7 (tetrahydro-2H-pyran-4-yl)ethyl)amino)but-2-enoate HN
(c)3...
_ -------------------- -------i 1 (R or S,Z)-methyl 2-(2-bromo-4-chloropheny1)-34(1- 0 i .....

(tetrahydro-2H-pyran-4-ypethyl)arnino)but-2-enoate HN Br COr'44' AI. F
VI
(R or S,Z)-methyl 2{2-bromo-4-fluoropheny1)-3-01-I Br 401 (tetrahydro-2H-pyran-4-ypethyl)amino)but-2-enoate 1N
CI).''' ao dish (R or S,Z)-methyl 2-(2-bromopheny1)-3-41- 0 la"
(tetrahydro-2H-thiopyran-4-yflethypamino)but-2- HN Br ..

enoate sraLi.
( )-(Z)-methyl 2-(2-bromopheny1)-3-01 -(3- 368 methyloxetan-3-ypethypamino)but-2-enoate HN
COC.0 [003041 Step 6: (R or S)-methyl 1-(1-(1-(tert-butoxycarbonyl)piperidin-4-yOethy1)-2-methyl-1H-indole-3-carboxylate:

MsOPdL.5 0 13r 1,4 y A 250 mL round bottom flask was charged with a magnetic stir bar, (R or S,Z)-tert-butyl 4-(1-(3-(2-bromophcny1)-4-methoxy-4-oxobut-2-en-2-yla.mino)ethyl)piperidine-1-carboxylatc (3.33 g, 6.92 nunol), RuPhos Prc-catalyst 11 (Chloro-(2-Dicyclohcxylphosphino-2',6'-diisopropoxy-1,1r-bipheny1)[2-(2-aminoethyl)phenyl]palladium(11) ¨ methyl-t-butyl ether adduct) (0.463 g, 0.553 mm.o1), dicyclohexyl(2',6'-diisopropoxybipheny1-2-yl)phosphine (0.387 g, 0.830 mmol), anhydrous 1,4-dioxane (27.7 ml, 6.92 mmol), and sodium methoxide (0.561 g,
10.38 mmol).
The reaction mixture was purged and back-filled with nitrogen and heated to 100 C with stirring overnight before being allowed to cool to it The reaction was diluted with ethyl acetate (¨ 100 ml) and the mixture was filtered through a bed of diatomaceous earth. The filtrate was pre-absorbed onto silica gel (-30g) and purified via silica gel chromatography (120 g) using ethyl a.cetate/hexanes (1:1) as eluent to afford the title compound (2.01 g, 4.77 mmol, 68.9 % yield).
1003051 The intermediates shown in the following table were prepared according to the general procedure outlined in Step 6 using the appropriate starting materials.
Name Structure I raiz.
I

Name Structure rniz.
\o (R or S)-methyl 2-methyl-I -(1-(tetrahydro-2H-pyran-4-yl)ethyl)-1H-indole-3-carboxylate --------- ----\
(R or S)-methyl 11 6-chloro-2-methyl-1-(1-(tetrahydro-2H-pyran-4-yl)ethyl)-1H-indole-3-carboxylate 0 o (R or S)-methyl 10 6-methoxy-2-methyl-1-(1-(tetrahydro-33'2 2H-pyran-4-ypethyl)- I H-indole-3-carboxylate 1 o \o (R or S)-methyl 6-fluoro-2-methy1-1-(1-(tetrahydro-2H-
11 3)0 pyran-4-y Dethyl)-1H-indole-3-carboxylate 0 (R or S)-methyl 2-methyl.- I -(1-(tetrahydro-2H- =

thiopyran-4-ypethyl)- I H-indole-3-carboxylate 0 ____________________________________________________________________ ( )-m ethyl 2-methy1-1-(1-(ox.etan-3-ypethyl)-1H-2'74 indole-3-carboxylate [003061 Step 7: (R or S)-2-methyl-1-(1-(tetrahydro-211-pyran-4-Aethyl)-1H-indole-3-carboxylic acid:
04"
NaOH
N = N

14 =
== = =
0 .1 Am,. =

A 1 L round bottom flask was charged with a magnetic stir bar, (R or S)-methyl 2-methy1-1-(1-(tetrahydro-2H-pyran-4-ypethyl)-1H-indole-3-carboxylate (11.60 g, 38.5 mmol), ethanol (96 ml, 38.5 mmol). and 6 N aqueous NaOH (64.1 ml, 385 mmol). The flask was fitted with a reflux condenser and heated to reflux for 6 h before being allowed to cool to rt. The volatiles were removed in vacuo and the resulting mixture was poured into 10% HCI (-300 mL).
A precipitate formed which was collected via vacuum filtration using a Buchner funnel. The filter cake was rinsed with an additional portion of water (-200 naL), collected, and dried under vacuum to afford the title compound (10.87 g, 35.9 mmol, 93 % yield) as an off-white solid.
1003071 The intein ediates shown in the following table were prepared according to the general procedure outlined in Step 7 using the appropriate starting materials.
Name Structure miz.
HO
(R or S)-2-methyl- I -(1-(tetrahydro-2H-pyran-4-yDethyD-I H-indole-3-carboxylic acid HO
(R or S)-6-chloro-2-methyl-1-(1-(tetrahydro-2H-pyran-4-yDethyl)-1H-indole-3-carboxylic acid HO
(R or S)-6-methoxy-2-methy1-1-(1-(tetrahydro-2H-pyran-4-yDethyl)-1H-indole-3-carboxylic acid HO
(R or S)-6-fluoro-2-methy1-1-(1-(tetrahydro-2H-pyran-4-yDethyl)-1H-indole-3-carboxylic acid Ds ,o (R or S)- 1-( 1-(1 ,1-dioxidotetrahydro-2H-thiopyran-4-yDethyl)-2-methyl-1H-indole-3-carboxylic acid ......

( )-2-methy1-1-(1-(oxelan-3-yDethyl)-1H-indole-3-HO
N--<0 274 carboxylic acid µr-C
(R or S)-2-methyl-6-(pyridin-3-y1)-1-(1-(tetrahydro-2 pyran-4-yDethyl)-11I-indole-3-carboxylic acid HO *

Name Structure miz (R or S)-2-methy1-6-(pyrazin-2-y1)-1-(1-(tetrahydro-2H- N 4 pyran-4-yl)ethyl)-1H-indole-3-carboxylic acid HO
\
0 ItisN
co (R or S)-2-metby1-1-(1-(tetrahydro-2H-pyran-4-ypethyl)-6-(thiazol-4-y0-1H-indole-3-carboxylic acid HO
# N.1 C) 1003081 Step 8: (R or S)-tert-butyl 4-(1-(3-(4-methoxy-6-methy1-2-oxo-1,2-dik5dropyridin-3-yl)methylearbamoy1)-2-methyl-1H-ind ol-1-yl)ethyl)piperidine-ea rboxylate (Compound 327).

,,keN
CO2H PF6' )-0-C4-HCI
HN''' I NH2 I ri -7( A 250 mL round bottom flask was charged with a magnetic stir bar, (R or S)-1-(1-(1-(tert-butoxycarbortyppiperidin-4-Aethyl)-2-methyl-1H-indole-3-carboxylic acid (1.950 g, 5.05 mmol), 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(1H)-one hydrochloride (2.065 g, 10.09 mmol), DMF (25.2 ml, 5.05 mmol), Hunig's base (3.52 ml, 20.18 mmol). The reaction mixture was cooled to 0 'C and COMU (2.16 g, 5.05 mmol) was added. The reaction was allowed to stir overnight to room temperature. The reaction mixture was diluted with water and extracted with Et0Ac. The combined organic extract was washed with brine, dried with MgSO4, filtered and conc, in vacuo to afford the crude material which was purified via silica gel chromatography (120 g) using MeOHlethyl acetate (1:5) as eluent to afford the title compound (1.86 g, 3.29 mmol, 65.3 % yield). LCMS 537 (M-1-1)" NMR
(400MHz ,DMSO-d6) 8 = 11.83 - 11.71 (m, 1 H), 7.80 (br. s., 1 II), 7.73 (d, J = 7.6 Hz, 1 H), 7.62 (d, J= 7.8 Hz, 1 El), 7.06 (td, = 7.1, 14.4 Flz, 2 H), 6.21 (s, 1 H), 4.32 (br. s., 2 H), 4.16 (br. s., 1 H), 4.02 (br.
s., 1 H), 3.85 (s, 3 H), 3.75 (br. s., I F1), 2.70 (br. s., 1 H), 2.58 (s, 3 H), 2.37 (br. s., I FE), 2.21 (s, 3 H), 1.90 (d, J= 12.9 Hz, III), 1.53 (d, J = 6.9 Hz, 3 H), 1.35 (s, 10 H), L21 (br. s., 1 H), 0.89 (d, J
= 8.7 Hz, 1 H),0.67 (d, = 11.8 Hz, I H).
[003091 The compounds shown in the following table were prepared according to the general procedure outlined in Step 8 using the appropriate starting materials. The structures of the compounds are shown in Figure 1.
Compound Name NMR rri/z Number (400MHz, DMSO-d6) 8 11.66 - 11.52 (m, 1 H), 7.80 - 7.67 (m, 2 H), 7.67 -(R or S)-1-(I-(1,1- 7.58 (m, 1 H), 7.16 - 7.02 (m, 2 H), 1 dioxidotctrahydro-2H-thiopyran-4- 6.15 (s, 1 H), 4.32 (d, J = 4.5 Hz, 2 305 1 yl)ethyl)-N-04-methoxy-6-methyl- H), 3.84 (s, 3 H), 3.24 - 3.06 (m, 2 H), 486 I 2-oxo-1,2-dihydropyridin-3- 2.91 - 2.77 (m, 1 H), 2.75 - 2.65 (m, 1 yOmethyl)-2-methyl-IH-indole-3- H), 2.60 (br. s., 3 H), 2.35 - 2.23 (m, 1 carboxamide H), 2.20 (s, 3 H), 1.93 - 1.76 (m, 2 H), 1 1.56 (d, J= 6.5 Hz, 4 H), 1.17- 1.03 1 (m, 2 H) (R or S)-tert-butyl 4-(1-(3-(((4,6-1 dimethy1-2-oxo-1,2-1 dihvdropyridin-3-yl)methyl)earbamoy1)-2-methyl-11H-indol- I -yl)ethyl)piperidine-1-carboxylate (R or S)-tert-butyl 44143404-(difluoromethoxy)-6-methy1-2-436 1 oxo-1,2-dihydropyridin-3-1 yOmethypcarbamoy1)-2-methyl-1H-indo1-1-ypethyl)piperidine-1-1 carboxylate (R or S)-tert-butyl 44143-0(4-! ethyl-6-methyl-2-oxo-1,2-1 dihvdropyridin-3-437 1 " 535 Amethypcarbamoy1)-2-methyl-1 1H-indo1-1-yl)ethyl)piperidine-1-1 carboxylate Compound Name H NMR rnz Number (400MHz, DMSO-d6) 8 11.60 (s, 1H), 7.73-7.62 (m, 3H), 7.60 (d, 2H) 7.07-7.05 (m, 2H), 6.15 (s, 1H) 4.33 (R or S)-N-((4-methoxy-6-methyl- (s, 1H), 4.21-4.11 (m, 1H), 3.92 2-oxo-12-dihydropyridin-3- (br.d., 1H), 3.65 (d, 1H), 3.34-3.32 298 yOmethyl)-2-methy1-1-(1- (m, 1H), 3.02 (t, 1H), 2.61 (s, 3H), 438 (tetrahydro-2F1-pyran-4-ypethyl)- 2.48-2.44 (m, 1H), 2.20 (s, 3H), 1.84-1H-i ndole-3-carboxamide 1.81 (m, 1H), 1.54 (d, 3H), 1.40-1.38 (m,12H), 1.25-1.22 (m, 1H), 1.08-1.04 (m, 1H), 0.86 (br. s., 1H), 0.58 (hr. d., 1H) (400MHz, DMSO-d6) 6 11.57 (bf.
s., 1 H), 7.75 - 7.67 (in, 2 H), 7.48 (d, J = 10.7 Hz, 1 H), 6.90 (t, J = 8.5 Hz, I (R or S)-6-fluoro-N-((4-methoxy- 1 H), 6.13 (s, 1 H), 4.29 (d, J = 4.5 6-methy1-2-oxo-1,2- Hz, 2 H), 4.12 (hr. s., 1 H), 3.94 -300 I dihydropyridin-3-yl)methyl)-2- 3.87 (m, 1 H), 3.83 (s, 3 H), 3.64 (dd, 456 methyl-1-(1-(tetrahydro-2H-pyran- J= 3.6. 10.9 Hz, 1 H), 3.35 (br. s.. 1 4-y1)ethyl)-1H-indole-3- H), 3.05 . (br. s., 1 H), 2.56 (s, 3 11.
), 1 carboxamide 2.45 - 2.37 (m, 1 H), 2.18 (s, 3 H), 1.81 (d, J= 12.7 Hz, 1 H), 1.50 (d, J
= 6.9 Hz, 3 H), 1.40 - 1.29 (m, 1 H), 1.11 - 0.99 (m, 1 H), 0.61 (br. s., 1 H) (400MHz, DMSO-d6) 8 11.57 (s, 1 H), 7.75 (s, 2 H), 7.66 (dõI = 8.9 Hz, (R or S)-6-chloro-N-((4-methoxy- 1 H), 7.08 (d, .1 = 8.5 Hz, 1 H), 6.14 (s, 1 H), 4.30 (d, J = 4.5 Hz, 2 H), 6-rnethy1-2-oxo-1,2-4.21 - 4.05 (m, 2H), 3.91 (d, J = 114 314 I dihydropyridin-3-Amethyl)-2-1 methy1-1-(1-(tetrahydro--)H-pyran- Hz, 1 H), 3.8 5 (s, 3 H), 3.65 (d, = 472 4-yDethyl)-1H-indole-3- 10.5 Hz, 1 H), 3.02 (t, 11.3 Hz, 1 H), 2.58 (s, 3 H), 246- 231 (m, 1 H), 1 carboxamide 2.19 (s, 3 H), 1.82 (d, J= 12.0 Hz, 1 H), 1.59 - 1.45 (m, 4 H), 1.44 - 1.29 (m, 1 H), 0.57 (d, J = 12.9 Hz, 1 H) Compound Name H NMR rnz Number (400MHz, DMSO-d6) 8 = 11.59 (s, 1 H), 7.67 - 7.59 (m, 2 H), 7.03 (s, 1 H), 6.75 - 6.68 (m, 1 H), 6.14 (s, 1 H), 1 (R or S)-6-methoxy-N-((4- 4.30 (d, J = 5.1 Hz, 2 H), 4.10 (dd, J
methoxy-6-methy1-2-oxo-1,2- = 7.5, 10.4 Hz, 1 H),3.91 (dd, J= 3.0, 321 1 dihydropyridin-3-yl)methy1)-2- 11.3 Hz, 1 H), 3.83 (s, 3 H), 3.80 - 468 methyl-1-(1-(tetrahydro-21I-pyran- 3.76 (m, 3 H), 3.68 - 3.60 (m, 1 H), 4-yl)ethyl)-1H-indole-3- 3.38 - 3.32 (m, 1 H), 3.10 - 3.00 (m, 1 1 carboxamide H), 2.56 (s, 3 H), 2.19 (s, 3 H), 1.83 (d, J = 12.7 Hz, 1 H), L55 - 1.43 (m, 1 4 H), 1.34 (br. s., 1 H), 1.10 - 0.96 (m, 1 H), 0.62 (d, J= 13.4 Hz, 1 H) (R or S)-N-((4-(difluoromethoxy)-6-methy1-2-oxo-1,2-335 I dihydropyridin-3-yl)methyl)-2-methy1-1-(1-(tetrahydro-2H-pyran-1 4-yl)ethy1)-11-1-i ndo le-3-carboxamide 1 (R or S)-N-((4,6-dimethy1-2-oxo-394 I 1,2-dihydropyridin-3-yOmethyl)-2-1 methyl -1 -(1 -(tetrahydro-2H-pyran- 422 4-yl)ethyl)-1H-i ndo le-3-carboxamide (400MHz, d6-DMS0) 8 11.59 (br. s., I1 1H), 7.72 (br. s., 2H), 7.05 (d, J =
( )-N-((4-Methoxy-6-methyl-2-1 =
oxo-1.2-dihydropyridin-3-(m, 1H), 4.64 (d, J = 6.24 Hz, 1H), 291 yl)methyl)-2-methy1-1-(1-(3-4.43 - 4.54 (m, 1H), 4.32 (d, J = 4.24 424 1 methyloxetan-3-ypethyl)-1H-Hz, 2H), 4.19 (d, J = 5.80 Hz, 1H), 1 indole-3-carboxamide 4.10 - 4.16 (m, 1H), 3.84 (s, 3H), 2.53 - 2.71 (m, 3H), 2.20 (s, 3H), 1.72 (d, J

1 = 6.91 Hz, 3H), 1.00 (s, 3H) (R or S)-6-chloro-N-((4,6-1 di methy1-2-oxo-1,2-442 I dihydropyridin-3-yOmethyl)-2-methyl-1-(1-(tetrahydro-2 H-pyran -1 4-yl)cthyl)-1 H-indole-3-carboxamide Compound 1 Name !El NMR rniz Number i 1 (400MHz, DMSO-d6) 8 11.54 (s, 1 H), 8.92 (br. s., 1 H), 8.53 (br. s., 1 H), 8.10 (d, J= 8.0 Hz, 1 H), 8.02 (s, 1 H), 7.91 - 7.82 (m, 1 H), 7.75 (d, J
r S)-N-((4 6-dimethy1-2- - " ' = 8 7 Hz 1 H) 7.54 - 7.40 (m 2 H) (R o,oxo 5.89 (s, 1 H), 4.39 - 4.27 (m, 2 H), 1 1,2-dihydropyridin-3-yl)methyl)-2-4.24 - 4.12 (m, 1 H), 3.93 (d, J = 7.6 499 413 I -1-methy1-6-(pyridin-3-y1) 1-I
Hz, 1 H), =3.66 (d, J = 7.4 Hz. 1 H), (tetrahydro-2H-pyran-4-ypethyl)- =
3.04 (t, J = 12.5 Hz, 1 H), 2.61 (s, 3 I 1H-indole-3-carboxamide 1 H), 2.48 - 2.37 (m, 1 H), 2.26 (s, 3 H), 2.12 (s, 3 H), 1.86 (d, J= 12.5 Hz, 1 H), 1.63 - 1.48 (m, 4 H), 1.46 - 1.32 (m, 1 H), 1.17 - 0.99 (m, 1 H), 0.65 (d,J- 12.7 Hz, 1 H) (R or S)-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-443 1 methyl-6-(pyrazin-2-y1)-1-(1- 500 (tetrahydro-2H-pyran-4-yDethyl)-1 1H-indole-3-carboxamide (R or S)-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-444 1 methy1-1-(1-(tetrahydro-2H-pyran- 505 4-yl)cthyl)-6-(thiazol-4-y1)-1H-indole-3-carboxamide [003.10] Step 9: (R or S)-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-y1)methy1)-2-methyl-1-(1-(piperidin-4-yDethyl)-1H-indolle-3-carboxamide hydrochloride (Compound 326).
(N HI
f HN I 0,11):01 HN N

(R or S) (Ft or S) A 250 mL round bottom flask was charged with a magnetic stir bar, (R or S)-tert-butyl ((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yOmethylcarbamoy1)-2-methyl-1H-indol-1-yDethyDpiperidine- 1 -carboxylate (Compound 327) (1.850 g, 3.45 mmol), Me0H
(13.79 ml, 3.45 mmol), and HC1 (2.59 ml, 10.34 mmol) (4 N in dioxane). The reaction was allowed to stir at rt for 6 h before being conc. in vac-uo to afford the title compound (1.65 g, 3.14 mmol, 91 % yield).
I,CMS 437 (M-Fl).
[003111 The compounds shown in the following table were prepared according to the general procedure outlined in Step 9 using the appropriate starting materials. The structures of the compounds arc shown in Figure 1.
Compound Name 111 NMR rnIz.
Number (400MHz, DMSO-d6) 6 11.61 (hr. s., 1 H), 8.52 (d, J = 10.3 Hz, 1 H), 8.14 (br. s., 1 H), 7.75 (d, J= 7.8 Hz, 1 H), 7.68 - 7.58 (m, 2 H), (R or S)-N-((4,6-7.15 - 7.03 (m., 2 5.90 (s, 1 H), 4.38 - 4.25 dimethy1-2-oxo-1,2-(m, 2 .11.), 4.25 - 4.14 (m, I li), 3.37 (d, J =
dihydropyridin-3-379 12.0 Hz, 1 H), 3.08 (dõ/ = 12.7 Hz, 1 H) 2.91 yl)m.ethyl.)-2-methyl.-1-(1- , 421 (piperidin-4-yDethyl)-1H- (d, J = 12.7 Hz, 1 H), 2.-73 - 2.61 (m., 2 H), indole-3-carboxamide 2.58 (s, 3 H), 2.27 (s, 3 H), 2.12 (s, 3 H.), 2.07 h ydrochloride s., 1 H), 1.56 (d, J = 6.9 Hz, 3 H), 1.46 (br. s., 1 H), 1.16 (d, .1 = 11.1 Hz, 1 H), 0.86 (d,.1 = 13.4 Hz, I H) (R or S)-N-((4-(difluoromethoxy)-6-methy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-2-methyl-1-(1-(piperidin-4-yDethyl)-1H-indole-3-carboxamide hydrochloride (R or S)-N-((4-ethyl-6-methy1-2-oxo-1,2-dihydropyridin-3-439 yOmethyl)-2-methyl-1-(1- 435 (piperidin-4-yDethyl)-1H-indole-3-carboxamide hydrochloride Compound NameH NMR 111/7, Number (400MHz, DMS0-4) 8 12.27 - 12.10 (m, 1 H), 11.96 - 11.72 (m, 1 H), 9.80 (br. s., 1 H).
9.19 (br. s.. 2 H), 7.89 - 7.67 (m, 2 H), 7.62 (R or S)-1-(1-(1-(azetidin-3-yl)piperidin-4-yl)ethyl)-(d, J= 7.6 Hz, 1 H), 7.09 (quin, J= 6.6 Hz, 2 N-((4,6-dimethy1-2-oxo-H), 5.99 (s, 1 H), 4.59 - 4.36 (m, 3 H), 4.24 -376 1,2-dih ydropyri din-3-3.95 (m, 2 FT), 3.48 (d, J= 13.2 Hz, 1 H), 3.17 (d, J= 12.0 Hz, 1 H), 2.87 (br. s., 1 H), 2.70 yl)methyl)-2-methyl-1 indole-3-carboxamide (br. s., 2 H), 2.58 (s, 3 H), 2.34 - 2.25 (m, 3 hydrochloride H), 2.19 - 2.10 (m, 3 H), 1.75 (d, J = 12.3 Hz, 1 H), 1.57 (d, J= 6.7 Hz, 3 H), 1.47 (d, J=
12.7 Hz, 2 H), 1.33- 1.21 (m, 2 H), 0.85 (d, J
= 13.6 Hz, 1 H) [003121 Step 10: (R or S)-isopropyl 4-(1-(34(4-methox-6-methy1-2-oxo-1,2-dihydropyridin-3-Amethylcarbamoy1)-2-methyl-111-indol-1-yl)ethyl)piperidine-1-carboxylate (Compound 346).
1;441.0H
o (1) I HCI I [41 Aka 0 0 o W
A 250 mL round bottom flask was charged with a magnetic stir bar, (R or S)-N-((4-metboxy-6-methy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl-1-(1-(piperidin-4-y1)ethy1)-1H-indole-3-carboxamide hydrochloride (0.467 g, 0.987 mmol) (Compound 326), DMF (2.468 ml, 0.987 mmol), THF (2.468 ml, 0.987 mmol), and N-ethyl-N-isopropylpropan-2-amine (0.638 g, 4.94 mmol). The reaction was cooled to 0 'C and isopropyl carbonochloridate (0.160 ml, 1.086 mmol) was added drop wise via syringe. The reaction was allowed to stir for 2 h to rt and was then treated with 5 N Li0F1 for 1 h to remove any acylated pyridone. This material was extracted with ethyl acetate, washed with brine, dried with MgSO4 and filtered and conc. in vacuo. The resulting material was purified via silica gel chromatography (50 g) using ethyl acetate/Me0H (5:1) as eluent to afford pure title compound as a pale yellow solid (0.300 g, 0.545 mmol, 55.2 % yield). LCMS 523 (M+1)+; NMR (DMSO-d6, 400 MHz) 8 11.59 (br. s., 1 H), 7.74 (d, J= 7.8 Hz, 1 H), 7.69 (t, J= 4.9 Hz, 1 H), 7.62 (d, J= 7.8 Hz, 1 H), 7.13 - 7.01 (m, 2 H), 6.15 (s, 1 H), 4.78 - 4.67(m., 1 H), 4.32 (d, J= 4.9 Hz, 2 H), 4.23-4.12 (m, 1 H), 4.12 -4.02 (m, 1 H), 3.84 (s, 3 H), 3.82 - 3.74 (m, 1 H), 2.79 - 2.66 (m, 1 H), 2.58 (s, 3 H), 2.46 - 2.34 (m, 2 H), 2.20 (s, 3 H), 1.96- 1.88(m, 1 H), 1.58- 1.46 (m, 4 H), 1.15 (d, J=
6.0 Hz, 6 H), 0.95 - 0.89 (m, 1 H), 0.74 - 0.65 (m, 1 H).
[003131 The compounds shown in the following table were prepared according to the general procedure outlined in Step 10 using the appropriate starting materials. The structures of the compounds are shown in Figure 1.
Compound Name NMR. Trilz Number (400MHz, DMSO-d6) = 11.59 (s, 1 H), 7.78 - 7.66 (m, 2 H), 7.64 -7.57 (m, 1 H), 7.06 (s, 2 H), 6.14 (s, (R or S)-N-44-methoxy-6-methyl-2-1 H) 4.31 (d, J= 4.9 Hz, 2 H), 4.25 oxo-1,2-dihydropyridin-3-- 4.15 (m., 1 H), 3.83 (s, 3 H), 3.63 yOmethyl)-2-methyl-1-(1 -(l -336 (s, 1 H), 3.40 - 3.33 (m, 1 H), 2.79 (methylsulfonyl)piperidin-4-(s, 3 H), 2.75 - 2.65 (m, 1 H), 2.60 yflethyl)-1H-indole-3-carboxamide (s, 3 H), 2.45 - 2.27 (m, 1 H), 2.19 (s, 3 H), 2.06 - 1.98 (m, 1 H), 1.55 (d, = 6.9 Hz, 3 H), 1.45 - 1.36 (m, 1 171), 1.28 - 1.18 (m, 1 H), 1.14 -1.03 (m, 1 H), 0.83 - 0.74 (m, 1 H) (400MHz, DMSO-d6) 8 11.58 (hr.
s., 1 H), 7.77 - 7.67 (m, 2 FT), 7.66 -(R or S)-1-(1-(1-(2-hydroxy-2- 7.60 (m, 1 H), 7.06 (s, 2 H), 6.14 (s, methylpropanoyl)piperidin-4- 1 H), 5.32 - 5.23 (m, 1 H), 4.31 (d, 337 yl)ethyl)-N-04-methoxy-6-methyl-2- J= 4.5 Hz, 2 H), 4.19 - 4.10 (m, oxo-1,2-d hydropyridi n-3- H), 3.83 (s, 3 H). 2.75 - 2.62 (m, 2 yl)methyl)-2-methyl- 1H-indole-3- H), 2.58 (s, 3 H), 2.19 (s, 4 H), 2.00 carboxamide - 1.90 (m, 2 H), 1.54 (d, J= 6.7 Hz, 3 H), 1.32 - 1.18 (m, 8 H), 0.87 -0.78 (m, 1 H), 0.77 - 0.67 (m, 1 H) (400MHz, DMS0-4) 8 = 11.59 (s, 1 H), 7.75 (d, J= 7.4 Hz, 1 H), 7.72 - 7.67 (m, 1 H), 7.64 (d, J= 8.0 Hz, 1 H), 7.14 - 7.01 (m, 2 H), 6.15 (s, (R or S)-1-(1-(1-isobutytylpiperid.in- 1 H), 4.58 - 4.46 (m, 1 H), 4.32 (d, 342 4-ypethyl)-N-04-methoxy-6-methyl- J= 4.9 Hz, 2 H), 4.09 - 3.99 (m, 1 2-oxo-1,2-dihydropyridin-3- H), 3.84 (s, 3 H), 3.81 - 3.72 (m, 1 yl)methyl)-2-methy1-1H-indole-3- H), 3.08 - 2.97 (m, I H), 2.92 - 2.81 carboxami de (m, 1 H), 2.78 - 2.65 (m, 3 H), 2.59 (br. s., 3 H), 2.20 (s, 3 H), 2.03 -1.90 (in, 1 H), 1.59- 1.47 (m, 4 H), 1.02 - 0.86 (m, 6 H), 0.78 -0.69 (m, 1H) Compound Name IH NMR mtz Number (400MHz, DMSO-d6) 8 12.02 -11.95(m, III), 7.74 (d, J= 8.0 Hz, 1 H), 7.66 - 7.57 (m, 2 H), 7.11 -7.00 (m, 2 H), 6.08 (s, 1 H), 4.32 (R or S)-N-((4-(difluoromethoxy)-6- (d, J= 4.5 Hz, 2 H), 4.18 (d, J=7.1 344 methyl-2-oxo-1,2-dihydropyridin-3- Hz, 1 H), 3.64 (d, J = 12.3 Hz, yl)meth y1)-2-meth y1-1-(1-(1 H), 3.36 (d, J= 12.0 Hz, 1 H), 2.79 551 (methylsul fonyl)piperi di n-4- (s, 3 II), 2.75 - 2.65 (m, 2 H), 2.58 yl)ethyl)-1H-indole-3-carboxamide (s, 3 H), 2.45 - 2.27 (m, 2 H), 2.20 (s, 3 H), 2.07- 1.98 (m, 1 H), 1.55 (d, J= 6.9 Hz, 3 H), 1.40 (d, J= 8.2 Hz, 1 H), 1.10 (d, J= 8.9 Hz, 1 H), 0.79 (d, J= 12.5 Hz, 1 H) (400MHz, DMSO-d6) 8 11.57 (s, 1 H), 7.75 (d, J = 8.0 Hz, 1 H), 7.69 (t, J= 5.0 Hz, 1 H), 7.62 (d, J= 7.4 Hz, 1 H), 7.06 (d, J= 7.1 Hz, 2 H), 6.15 (s, 1 H), 4.32 (d, J= 5.1 Hz, 2 (R or S)-1-(1-(1- H), 4.25 - 4.15 (m, 1 H), 3.84 (s, 3 345 (ethy1su1fony1)piperidin-4-y1)ethy1)- H), 3.73 - 3.65 (m, 1 H), 3.45 - 3.36 N-((4-methoxy-6-rnethy1-2-oxo-1,2- (m, 1 H), 3.02 - 2.93 (m, J = 7.8 529 dihydropyridin-3-Amethyl)-2- Hz, 2 H), 2.87 - 2.77 (m, 1 H), 2.75 methyl-1H-indole-3-carboxamidc - 2.66 (m, 1 H), 2.60 (s, 3 H), 2.42 -2.30 (m, 1 H), 2.20 (s, 3 H), 2.06 -1.97 (m, 1 H), 1.58 - 1.48 (m, 4 H), 1.42- 1.31 (m, I H), 1.17 (t, J= 7.5 Hz, 3 H), 1.13- 1.00 (m, 1 H),0.83 - 0.73 (m, 1 171) (400MHz, DMSO-d6) 8 11.59 (s, 1H), 7.76-7.69 (m, 2H), 7.62 (d, 1H), 7.10-7.03 (m, 2H), 6.15 (s, (R or S)-1-(1-(4-1H), 4.32 (d, 2H), 4.29-4.26 (m, 355 (isopropylsulfonyl)cyclohexyl)ethy1)-2H), 3.84 (s, 3H), 3.72 (br. d.' 1H)' 543 N-((4-methoxy-6-methy1-2-oxo-1,2-3A5 (br. d., 1H), 3.26 (ft, 1H), 2.91 dihydropyridin-3-yOmethyl)-2-(dt. 1H), 2.60 (s, 3H), 2.20 (s, 3H), In ethy1-1 FI-indole-3-carboxam ide 1.97 (br. d., 1H), 1.54 (d, 3H), 1.35-1.24 (m, 2H), 1.18 (d, 3H), 1.16 (d, 3H), 1.05-0.78 (m, 2H) Compound Name IH NMR mtz Number (400MHz, DMSO-d6) 6 11.60 (br.s., 1H), 7.75-7.60 (m, (R or S)-isobutyl. 4-(1-(3-((4-3H), 7.10-7.03 (m, 2H), 6.15 (s, methoxy-6-methy1-2-oxo-1,2-1H) 4.33 (d, 1H), 4.13-4.06 (m, 357 dihydropyridin-3-1H), 3.84 (s, 3H), 3.74 (d. 1H), 537 yl)methyl)carbamoy1)-2-methyl-lii-2.80-2.60 (m 3H) 2.58 (sõ 1H), indo1-1-ypethyl)piperidine-1- , 5 2.50-2.42 (m, 2H), 1.96-1.90 (m, carboxylate 1H), 1.54 (d, 3H), 1.25-1.22 (m, 1H), 0.98-0.72 (m, 611) (400MHz, DMSO-d6) 8 11.59 (s, 1 H), 7.78 - 7.71 (m, 1 H), 7.66 - 7.57 (m, 2 H), 7.07 (s, 2 H), 5.89 (s, 1 H), 4.32 (s, 2 H), 4.25 - 4.15 (m, 1 (R or S)-N-((4,6-dimethy1-2-oxo-1,2- H), 3.65 -3.59 (m, 1 H), 3.19 -3.10 368 dihydropyridin-3-Amethyl)-1-(1-(1- (m, 1 H), 2.98 (d, J= 7.4 Hz, 2 H), 513 (ethylsulfonyppiperidin-4-yl)ethyl)- 2.87 - 2.77 (m, 1 H), 2.72 - 2.65 (m, 2-methyl-1H-indole-3-carboxamide 1 H), 2.58 (s, 3 H), 2.27 (s, 3 H), 2.12 (s, 3 H), 1.55 (d, J= 6.9 Hz, 4 H), 1.42- 1.33 (m, 2 H), 1.17 (t, J=
7.4 Hz, 3 H), 1.12 - 1.00 (in, 1 H), 0.84 - 0.74 (m, 1 H) (400MHz, DMSO-d6) 6 11.60 (s, 1 14), 7.75 (d, .1= 7.1 Hz, 1 H), 7.65 -7.58 (in, 2 H), 7.12 - 7.02 (m, 2 FI), .89 (R or S)-N-((4,6-dimethy1-2-oxo-1,2-(s,1 H),4.38 - 4.25 (m, 2 H), 4.20 dihydropyridin-3-yl)methyl)-2- .
382 2.80 (s, 3 H), 2.76 - 2.67 =(m, 2 49, II), methy1-1-(1-(1-2.59 (s, 3H), 246 - 2.31 (m, 2H)' (metilylsulfonyl)piperidin-4-2.27 (s, 3 H), 2.12 (s, 3 H), 1.55 (d, yl)ethyl)-1H-indole-3-carboxarnide .
.1= 6.9 Hz, 3 H), 1.51 (br. s., 1 H), 1.47- 1.34(m, 1 H), 1.29- 1.21 (m, 1 H), 1.17 - 1.04 (m, 1 H), 0.80 (d, J= 12.9 Hz, 1 H) [003141 Example 27. Synthesis of (R or S)-1-(1-(1-isopropylpiperldivi-4-N 1)etliv1)-N-((4-methoxy-6-ntethyl-2-oxo-1,2-diltydropyridin-3-yl)mcffiv1)-2-methyl-1 carboxamide (Compound 358) oy..0 0 Na*
=
Nr.011 (I) i I H I H ITc..õ,..N
HN N
0 0 o A 25 mL vial was charged with a magnetic stir bar, (R or S)-N44-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl-1-(1-(piperidin-4-ypethyl)-1H-indole-3-carboxamide hydrochloride (Compound 326), THF (2.114 mi., 0.211 mmol), propan-2-one (0.06.1 g, 1.057 mmol), and sodium triacetoxyborohydrid.e (0.224 g, 1.057 mmol). The reaction was allowed to stir at rt for 12 h. The reaction was inverse quenched onto sat'd aqueous NaHCO3, extracted with ethyl acetate and conc. in vacuo. The resulting material was treated with 10 mi, 7 N
ammonia in Me0H and was cone in vacuo to yield material which was purified via silica gel chromatography (10g) using DCM/Me0H/NH4OH (90:1:0.1) as eluent to afford 33 mg, (0.065 mmol, 31.0 % yield) of the title compound as a white solid.). LCMS 479 (M-1-1).'.; 11-1NMR.
(DMSO-d6, 400 MHz) 8 11.59 (s., 1H), 7.64-- 7.82 (m, 2H), 7.59 (d, 1H), 6.95 7.17 (m., 2H), 6.15 (s, 1H), 4.32 (d, 2 H), 4.04 - 4.24 (m, III), 3.84 (a, 311), 2.77 - 2.93 ( , 2H), 2.68 (8, 111), 2.60 (a, 3H), 2.20 (a, 3 H), 2.08 --2.15 (j.t, 1H), 1.92 (8, 111), 1.83 (13p. a., 1H), 1.54 (8, 3H), 1.27 1.43 ( , 2H), 0.91 (c, 6H), 0.71-0.67 (IA, 2H).
[003151 The compounds shown in the following table were prepared according to the general procedure outlined in this Example using the appropriate starting materials.
The structures of the compounds are shown in Figure 1.
Compound Name 1H NMR. miz Number (400MHz, DMSO-d6) 8 11.58 (s, 1 H), 7.76 -7.65 (m, 2 H), 7.59 (d, = 7.8 Hz, 1 H), 7.10 (R or S)-N-((4-methoxy- - 6.99 (in, 2 H), 6.14 (s, 1 H), 4.49 (t, J = 6.4 6-methyl-2-oxo-1,2- Hz, 1 H), 4.43 (t, J 6.5 Hz, 1 H), 4.37 (t, =
dihydropyridin-3- 6.1 Hz, 1 H), 4.34 - 4.28 (m, 3 H), 4.21 -4.10 341 yl)methyl)-2-methyl-1-(1- (m, 1 H), 3.83 (s, 3 If), 3.30 - 3.23 (m, 1 H.), 493 I (1-(oxetan-3-yl)piperidin- 2.75 (br. s., 1 H), 2.71 - 2.64 (m, 1 H), 2.60 4-yl)ethyl)-1H-indole-3- (s, 3 H), 2.19 (s, 4 H), 1.90 (br. s., 1 H), 1.75 carboxamide (hr. s., 1 H), 1.53 (d, = 6.9 Hz, 3 H), 1.42 (hr. s., 2 H), 1.11 - 0.98 (m, 1 H), 0.72 - 0.63 (m, 1 H.) Compound Name 1E1 NMR
Number (400MHz, DMSO-d6) 8 11.58 (s, 1 H), 7.76 -(R or S)-N-((4-methoxy- 7.65 (m, 2 H), 7.59 (d, J= 7.6 Hz, 1 H), 7.11 6-methyl-2-oxo-1,2- -6.99 (m, 2 H), 6.14 (s, 1 H),4.31 (d, J = 5.1 343 1 ' dihvdropyridin-3- Hz, 2 H), 4.13 (br. s., 1 H), 3.83 (s, 3 H), 2.83 yl)methyI)-2-methyl-1-(1- (d. J= 10.0 Hz, 1 H), 2.61 - 2.52 (m, 5 H), 451 (I-methylpiperidin-4- 2.19 (s, 3 H), 2.09 (s, 4 H), 1.88 (d, J= 10.7 ypethvi)-1H-indole-3- Hz, 2 H), 1.53 (d, J = 6.7 Hz, 3 H), 1.34 (br.
carboxamide s., 1 H), 1.02 (d, J = 8.2 Hz, 1 H), 0.66 (br.
s., 1 H) (400MHz ,DMSO-d6) 8 11.59 (s, 1 H), 7.77 (R
7.66 (in, 2 H), 7.60 (d, J= 7.8 Hz, 1 11), 7.12 6-methyl-2-oxo-12-or S)-N-((4 -me th oxy-- 7.01 (m, 2 H), 6.15 (s, 1 H), 4.32 (d, J= 4.9 , Hz, 2 H), 4.13 (d,J= 7.1 Hz, 1 H), 3.85 (s, 3 dihydropyridin-3-õ14õ11 359 vil. H), 3.36 (t, J = 5.9 Hz, 2 H), 3.19 (s, 3 H), 2 94 (d, J = 10.5 Hz, 1 H),2.71 - 2.56 (m, 5 methoxyethyppiperidin- =
H), 2.43 - 2.32 (m, 2 H), 2.24 - 2.12 (m, 4 H), 1.54 (d, J = 6.9 Hz, 4 H), 1.39- 1.27 (m, 2 indole-3-earboxamide H), 1.02 (d, J = 8.7 Hz, 1 H), 0.65 (d, J = 12.7 Hz, 1 H) (400MHz, DMSO-d6) 8 11.79 - 11.45 (m, 1 H), 7.78 - 7.65 (m, 2 H), 7.59 (d, .1= 7.8 Hz, I (R or S)-1-(1-(1-1 11), 7.14 - 6.99 (m, 2 H), 6.15 (s, 1 H), 4.32 ethylpiperidin-4-yl)ethyl)-(d, J = 4.9 Hz, 2 H), 4.20 - 4.08 (m, 1 H), 360 N-04-methoxy-6-methyl-3.84 (s, 3 H), 2.98 - 2.89 (m, 1 H),2.71 -2.61 465 2-oxo-1,2-dihydropyri (m, 2 H), 2.59 (s, 3 H), 2.27 - (m, 2 H), 3-yl)m=ethyl)-2-methyl-2.20 (s, 3 H), 1.94 - 1.80 (m, H), 1.54 (sõ 4 1H-indo le-3-earbox amide H), 1.38 - 1.28 (m, 1 H), 1.06 - 0.98 (m, 1 H), 0.93 (t, J = 7.1 Hz, 3 H), 0.71 - 0.63 (m, I H) (400MHz, DMSO-d6) 8 11.59 (br. s., 1 H), (R or S)-ethyI 2-(4-(1-(3- 7.81 - 7.65 (m, 2 1-1), 7.60 (d, 1 = 7.4 Hz, 1 (04-methoxy-6-methy1-2- H), 7.16 - 6.98 (m, 2 H), 6.15 (s, 1 H), 4.32 i (d, .1 = 4.9 Hz, 2 H), 4.23 - 4.11 (m, 1 H), oxo-1' " 2-dihydropyridin-3-363 4.04 (q, =7.0 Hz, 2 H), 3.84 (s, 3 H), 2.95 -ypmethypearbamoy1)-2- 523 2.86 (m, 1 H), 2.60(s, 5H1, 2.20 (s, 4 H), methyl-I1I-indo1-1-1.94 - 1.79 (m, 2 H), 1.54 (d, 1= 6.9 Hz, 4 ypethyl)piperidin-1-H), 1.41- 1.32 (m, 1 H), 1.15 (t,1=7.1 Hz, 3 yl)acetate H), 1.04 (d, J = 6.0 Hz, 2 H), 0.71 - 0.61 (m, 1H) Compound Name 1H NMR miz Number (400MHz, DMSO-d6) 8 11.63 (s, 1 H), 7.74 (R or S)-N-04-ethy1-6- (d, J = 7.6 Hz, 1 H), 7.65 - 7.56 (m, 2 H), methy1-2-oxo-1,2- 7.12 -7.01 (m, 2 H), 5.94 (s, 1 H), 4.34 (t, J=
366 ' dihvdropyridin-3- 5.1 Hz, 2 H), 4.19 - 4.09 (m, 1 H), 2.88 (br.
vpmethy1)-2-methyl-1-(1- s., 1 H), 2.71 - 2.56 (m, 6 H), 2.14 (s, 7 H), 449 (1-methy !piped di n-4- 1.91 (d, J =
12.5 Hz, 1 H), 1.54 (d, J = 6.9 I ypethyl)-1H-indole-3- Hz, 4 H), 1.41 - 1.31 (m. 2 H), 1.14 (t, J = 7.6 carboxamide Hz, 3 H), 1.05 (d, J = 9.1 Hz, 1 H), 0.68 (d, J
= 12.7 Hz, 1 H) (400MHz, DMSO-d6) 8 11.59 (s, 1 H), 7.74 (R or S)-N-((4,6-(d, J = 6.9 Hz, 1 H), 7.65 - 7.56 (m, 2 H), dimethy1-2-oxo-1,2- 7.12 - 7.01 (m, 2 H), 5.89 (s, 1 H), 4.38 - 4.25 dihydropyridin-3- (m, 2 H), 4.20 - 4.09 (m, 1 H), 2.95 (br. s., 367 1 yl)methyl)-2-methy1-1-(1- H), 2.68 (br. s., 2 H), 2.58 (s, 3 H), 2.27 (s, 3 .. 435 (1-methylpiperidin-4- H), 2.21 (br.
s., 3 H), 2.12 (s, 3 H), 1.94 (d, J
I ypethyl)-1H-indole-3- = 13.8 Hz, 1 H), 1.54 (d, J = 6.9 Hz, 4 H), carboxamide 1.44- 1.31 (m, 2 H), 1.07 (d, J = 12.5 Hz, 1 H), 0.71 (d, J = 13.2 Hz, 1 H) (400MHz, DMS0-4) 8 11.59 (br. s., 1 H), 7.73 (d, = 7.6 Hz, 1 H), 7.65 - 7.55 (m, 2 (R or S)-N-((4,6-H), 7.12 - 7.00 (m, 2 H), 5.89 (s, 1 H), 4.53 -dimethy1-2-oxo-1,2- 4.48 (m, 1 H), 4.47 - 4.42 (tn. 1 H), 4.38 (s, 1 dih ydropyridi n-3- H), 4.31 (t, J= 5.2 Hz, 3 H), 4.21 - 4.10 (m, 1 375 I yOmethyl)-2-methyl-1-(1- H), 3.31 -3.24 (m, 2 H), 2.81 - 2.64 (m, 2 H), 477 (1-(oxetan-3-yl)piperidin- 2.59 (s, 3 H), 2.26 (s, 3 H), 2.23 - 2.16 (m, 1 I 4-ypethyl)-1H-indole-3- H), 2.12 (s, 3 H), 1.98 - 1.85 (m, 1 H), 1.81 -carboxamide 1.70 (m, 1 H), 1.54 (d, .1= 6.9 Hz, 3 H), 1.51 - 1.22 (m, 1 H), 1.12 - 0.96 (m, 2 H), 0.73 -0.64 (m, 1 H) (400 MHz, DMSO-d6) 8 11.63 (br. s., 1 El), 7.74 (d, J - 7.36 Hz, 1 H), 7.60 (d, I = 8.47 Hz, 2 111), 7.06 (quin, = 7.13 Hz, 3 IT), 5.94 (s, 1 11), 4.51 (t, J = 6.47 Hz, 1 Fl), 4.46 (t, J=
(R or S)-N-04-ethy1-6- 6.35 Hz, 1 H.), 4.40 (t, I = 6.13 Hz, 1 H), 4.37 I methyl-2-oxo- 1,2- - 4.30 (m, 2 H), 4.28 - 4.11 (m, 1 H), 3.57 (s, 380 dihydropyridin-3- 1 H), 3.34 (br. s., 2 H), 2.81 (d, = 10.70 Hz, YOmethyl)-2-methyl-1-(1- 1 H), 2.67 (d, J = 14.94 Hz, 1 H), 2.64 - 2.57 491 (1-(oxetan-3-yl)piperidin- (m, 4 H), 2.21 (dõ1 = 10.93 Hz, 1 H), 2.14 (s, 4-ypethyl)-1H-indole-3- 3 H), 1.93 (d, .J= 12.49 Hz, 1 H), 1.83 (t, J =
carboxamide 11.37 Hz, 1 H), 1.54 (d, I = 6.91 Hz, 3 H), 1.37 (d, J = 10.48 Hz, 1 H), 1.25 (q, = 6.91 Hz, 1 H), 1.14 (t, 1' 7.58 Hz, 3 El), 1.06 (d, J
= 9.81 Hz, 1 H), 0.70 (d, J = 12.49 Hz, 1 H) Compound Name 1H NMR miz Number 114 NMR (400 MHz, DMSO-d6) 8 11.60 (br.
s., 1 H), 7.74 (d, J = 7.13 Hz, 1 H), 7.66 -(R or S)-N-((4,6-7.50 (m, 2 H), 7.15 - 6.99 (m, 2 H), 5.89 (s, 1 dimethy1-2-oxo-1,2- H), 4.40 -4.24 (m, 2 H), 4.21 - 4.07 (m, 1 H), dihydropyridin-3- 3.95 - 3.78 (m, 2 H), 3.57 (s, 1 H), 3.32 - 3.17 381 yl)methyl)-2-methyl-1-(1- (m, 3 H), 2.68 (br. s., 1 H), 2.58 (s, 3 H), 2.33 505 (1-(tetrahydro-2H-pyran- (br. s., 2 H), 2.27 (s, 3 H), 2.12 (s, 3 H), 2.00 4-yl)piperidin-4-yl)ethyl)- - 1.88 (m, 2 H), 1.75 (d, J = 12.04 Hz, 1 H), 1H-indole-3-carboxamide 1.62 (br. s., 2 H), 1.54 (d, J = 6.91 Hz, 3 H), 1.46- 1.30 (m, 2 H), 1.01 (br. s., 1 H), 0.72 (br. s., 1 H) (R or S)-tert-butyl 344-(1-(34(4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-440 I yOmethyl)carbamoy1)-2- 576 methyl-1H-indo1-1-yflethyl)piperidin-1-____01 Dazetidine-l-carboxylate (400MHz, DMSO-d6) 6 11.63 - 11.56 (m, 1 N 0 (R 6 dimeth H)' 7.76 -7.70 (m, 1 H), 7.64 - 7.55 (m, 2 H), or S ) 7.05 (s, 2 H), 5.89 (s, 1 H), 4.56 (s, 4 H), 4.31 dihydro ridin-3-y1-2-oxo-1,2- , (s, 2 H), 4.19 - 4.09 (m, 1 H), 3.36 (d, - 4.9 py 37 1 y1)methyl)-2-methy1-1-(1- Hz, 1 H), 2.77 - 2.56 (m, 5 H), 2.26 (s, 3 H), (1-(1-methylazetidin-3-2.18 (s, 3 H), 2.12 (s, 3 H), 1.94 - 1.85 (m, 1 yl)pi ridin-4-ypethyl H), 1.78 - 1.67 (m, 1 H), 1.53 (d, J= 6.9 Hz, 1H-ind i pe)-3 H), 1.50 - 1.45 (m, 1 H), 1.44 - 1.22 (m, 2 o e e-3-carlx)xamid H), 1.07 - 0.93 (m, 1 H),0.71 -0.61 (m, 1 H) [003161 Example 28. Synthesis of (R or S)-1-(1-(1-(2-111noroethyl)piwridi hettim1)-N-04-rnedloxv-6-methyl-2-oxo-I,2-diltvdropyridio-3-vinuet itµ1)-2-ntei ilµ1-11kndok-3-earboNamide (Conwound 356):

N
HCE I:" I M Ala HN N
vir A 25 naL, vial was charged with a magnetic stir bar, (R or S)-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(piperidin-4-ypethyl)-1H-indole-3-carboxamide hydrochloride (Compound 326) (0.062 g, 0.131 mmol), K2CO3 (0.072 g, 0.524 mmol), MeCN
(0.655 ml, 0.131 mmol), DMF (0.262 ml, 0.131 mmol) and 1-bromo-2-fluoroethane (0.020 ml, 0.262 mmol). The reaction was capped and heated to 82 C with stirring for 4 h. The reaction was allowed to cool to rt, filtered, and the filtrate was pre-absorbed onto silica gel (12 g). The material was purified via SiO2 chromatography (25 g) using DCM/Me0H/Et3N
(85:15:0.5) as eluent to afford the title compound as an off white solid (30 mg, 0.059 mmol, 45.1 % yield). ).
LCMS 483 (M+1)' ; NMR (DMSO-d6, 400 MHz) 8 11.59 (s, 1H), 7.75-7.68 (m, 2H), 7.60 (d, 1 H) 7.09-7.03 (in, 2H), 6.15 (s, 1H) 4.53-4.51 (m, 1H), 4.42-4.39 (m, 1H), 4.32 (d, 2H), 4.24-4.2 (m, 1H), 3.84 (s, 3H), 2.98 (br. d., 1H), 2.70-2.49 (m, 4H), 2.60 (s, 3H), 2.20 (s, 3H), 2.01 (dt, 1H), 1.92-1.90 (m, 1H), L75-1.71 (m, 1H), 1.54 (d, 3H), 1.38-1.36 (m, 1H), 1.02-0.98 (m, 1H), 0.7-0.66 (br. d., 1H).
1.00317] The compounds shown in the following table were prepared according to the general procedure outlined in this Example using the appropriate starting materials.
The structures of the compounds are shown in Figure 1.
Compound Name 111 NMR ink Number (R or S)-1-(1-(1-(2,2-(400 MHz, DMSO-d6) 8 = 11.60 (br. s., 1 H), difluoroethyl)pipe n.din-7.77 - 7.66 (m, 2 H), 7.60 (d, J = 7.6 Hz, 1 H), 7.14- 7.00 (m, 2 H), 6.15 (s, 1 H), 6.06 (t, J =
4-yl)ethyl)-N-04-55.7 Hz, 1 H), 4.32 (d, J = 4.9 Hz, 2 4.15 362 methoxy-6-methyl-2-oxo-1,2-dihydropyridin-(br. s., 1 H), 3.84 (s, 3 H), 3.03 - 2.93 (in, 2 H), 501 2.73 - 2.62 (m, 3 H), 2.60 (s, 3 H), 2.26 - 2.10 3-yl)methyl)-2-methyl-(m, 4 H), 1.93- 1.79 (m, 1 H), 1.59 - 1.46 (m, 4 1H-indole-3-carboxamide H), 1.41 - 1.29 (m, 1 H), 1.11 - 0.97 (m, 1 H), 0.67 (br. s., 1 H) (R or S)-N-((4-methoxy-(400MHz' DMSO-d6) 8 = 11.60 (br. s., 1 H), I 6-methy1-2-oxo-1,2-7.78 - 7.66 (m, 2 H), 7.60 (d, J = 8.2 Hz, 1 H), 7.13 - 7.00 (in, 2 H), 6.15 (s, 1 H), 4.32 (d, J=
dihydropyridin-3-4.9 Hz, 2 H), 4.22 - 4.09 (m, 1 H), 3.84 (s, 3 H), 378 ypincthyl)-2-methyl-1-(1-(1-(3,3,3-3.03 - 2.91 (m, 1 H), 2.73 - 2.64 (m, 1 H), 2.60 533 trifluoropropyl)piperidin-(s, 3 H), 2.48 - 2.31 (m, 5 H), 2.20 (s, 3 H), 2.01 4-yl)ethyl)-1H-indole-3-- 1.85 (m, 2 H), 1.58 - 1.46 (m, 4 H), 1.36- 1.29 (m, 1 H), 1.08 - 0.98 (m, 1 H), 0.73 - 0.62 (m, 1 carboxamide H) Compound Name NMR rn/z Number (500MHz, DMSO-d6) 8 = 11.59 (s, 1 H), 7.74 (R or S)-N-04-methoxy-(d, J = 7.6 Hz, 11), 7.71 - 7.66 (m, 1 H), 7.61 6-methyl-2-oxo-1,2-(d, J = 7.8 Hz, 1 H), 7.13 - 7.01 (m, 2 H), 6.15 dihydropyridin-3- I
(s, 1 H), 4.32 (d, J = 4.9 Hz, 2 H), 4.22 - 4.12 365 yl)methyl)-2-methyl-1-(m, 1 H), 3.84 (s, 3 H), 3.15 - 2.95 (m, 3 H), 1 (14142,22-2.75 - 2.66 (m, 1 H), 2.60 (s, 3 H), 2.39 - 2.31 519 trifluoroethyppiperidin-, I I
(m, 1 H), 2.20 (s, 3 H), 2.05 - 1.98 (m, 1 H), 1.92- 1.84 (m, 1 H), 1.56- 1.46 (m, 4 H), 1.42-I carboxamide 4-yl)ethyl)-1H-indole-3-1.32 (m, 1 H), 1.11 - 1.01 (m, I H), 0.69 - 0.62 (m, I H) (400MHz ,DMSO-d6) 8 = 11.59 (s, 1 H), 7.73 (d, J = 7.8 Hz, 1 H), 7.65 - 7.55 (m, 2 H), 7.12 -I (R or S)-1-(1-(1-(22-difluoroethyl)piperidin-7.00 (in, 2 H), 6.22 - 5.9441 .. 10 (m, 1 H), 5.89 (s, 1 4-yl)ethyl)-N-((4,6-I
H), 4.36 - 4.25 (m, 2 H), 4.20 - 4.09 (m, 1 H), dimethy1-2-oxo-1,2-3.01 - 2.93 (m, 1 H), 2.72 - 2.59 (m, 3 H), 2.58 I dihydropyridin-3-I yl)methyl)-2-methyl-1H-(s, 3 H), 2.26 (s, 3 H), 2.21 -2.13 (m, 2 H), 2.12 (s, 3 H), 1.92 - 1.79 (m, 2 H), 1.53 (s, 4 H), 1.41 indole-3-carboxamide - 1.29 (m, 1 H), 1.10- 0.97 (m, 1 H), 0.70- 0.59 (m, 1 F1) [003181 Example 29. Synthesis of (R or S)-N-((1-methoxy-6-methy1-2-oxo-1.2-dihydropyridin-3-v1)methyl)-2-methyl-1-(1-(1-(pyrimidin-2-v1)piperidin-4-v1)ethyl)-111-indole-3-carboxamide (Compound 361).
14)--CP*1 N CE3C; =-"( _a \t4 1.1 N
" IrLs =
To a re-sealable vial was added 2-chloropytimidine (185 mg, 1.611 mmol), (R or S)-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-Amethyl)-2-methyl-1-(1-(piperidin-4-ypethyl)-1H-indole-3-carboxamide hydrochloride (508 mg, 1.074 mtnol) (Compound 326), and Et0H (8 mL). To this solution was added Et3N (449 Al, 3.22 mmol). The vial was sealed and heated to 100 C overnight. The solution was allowed to cool to room temperature and concentrated in vacuo. The crude residue was purified via silica gel chromatography (hexanes:
(3:2 DCM:IPA)) to afford the title compound as a solid (357 mg, 0.694 rnmol, 64.6 % yield).
LCMS 515 (M-Flr;
NMR (DMSO-d6, 400 MHz) 8 11.60 (s, 1 H), 8.30 (d, J= 4.7 Hz, 2 H), 7.76 (d, J=
7.6 Hz, 1 H), 7.73- 7.64(m, 2 H), 7.14 - 7.01 (m, 2 H), 6.55 (t, I = 4.7 Hz, 1 H), 6.15 (s, 1 H),4.84 -4.75 (m, 1 H), 4.57 -4.47 (m, 1 H), 4.33 (d, J= 4.2 Hz, 2 H), 4.22 -4.11 (m, 1 H), 3.84 (s, 3 H), 2.92 -2.81 (m, 1 H), 2.63 - 2.52 (m, 4 H), 2.20 (s, 3 H), 2.05 - 1.94(m, 1 H), 1.61-1.49 (m, 4 H), 1.34- 1.21 (m, 1 H), 1.04- 0.91 (m, 1 FT), 0.83 -0.75 (m, I H).
[003191 The compound shown in the following table was prepared according to the general procedure outlined in this Example using the appropriate starting materials.
The structure of the compound is shown in Figure I.
Compound Name IFI NMR miz Number (R or S)-N-((4-methoxy-6-methyl-(400MHz, DMSO-d6) 8 11.60 (br. s., 1 H), 8.06 (d, J = 3.6 Hz, 1 H), 7.82 - 7.62 (m, 3 H), 7.51 -2-oxo-1,2-dihydropyridin-3- 7'39 (m' 1 H), 7.17 - 6.98 (m, 2 H), 6.75 (d, J =
8.5 Hz, 1 H), 6.61 - 6.49 (m, 1 H), 6.15 (s, 1 H), 373 yl)methyl)-2- methyl-1-(1-(1-4.49 - 4.38 (m, 1 H), 4.33 (d, J = 3.8 Hz, 2 H), 514 (pyridin-2-4.24 - 4.03 (m, 2 H), 3.85 (s, 3 H), 2.90 - 2.70 (in, yl)piperidin-4-2 H), 2.58 (s, 3 H), 2.20 (s, 3 H), 2.06 - 1.91 (m, 1 ypethyl)- I H-H), 1.63 - 1.47 (m, 4 H), 1.40 - 1.27 (m, 1 H), 1.07 indole-3-- 0.94 (m, 1 H), 0.82 - 0.72 (m, 1 H) carboxamide .
[003201 Example 30. Synthesis of (R or S)-1-f 1 4 1-(2-its droliA eth v1)91 neridi 11-4-µ
iliethyll)-N4(4-methoxv-6-methyl-2-oxo-1 ,2-dihvdropyrid i n-3-vi}rii e I h v1)-2-m et h v1-1 1-1-in d o le-3-carboxamide (Compound 347).
0 lr-0H

......r..../. ______________ Nix ,=-=I . Niik HN r4 iit, 0 . 0 -To a sealed tube charged with a magnetic stir bar was added (R or S)-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(piperidin-4-y1)ethyl)-1H-indole-3-carboxamide (Compound 326) (0.1 g, 0.229 mmol) was added DCM (3 mL) and the reaction cooled to 0 'C. To the cooled reaction mixture was added oxirane which was condensed into the reaction vial (-1 mL). The reaction was allowed to stir to it over 4 h and was then conc. in vacuo to afford the crude material which was purified via silica gel chromatography (12 g) using ethyl acetate/Me0H (4:1) as eluent to afford the title compound as a white solid (50 mg). LCMS

481 (M+1)- (DMSO-d6, 400 MHz) 8 ) 8 11.58 (s, 1 H), 7.77 - 7.65 (m, 2 H), 7.59 (d, 7.8 Hz, 1 H),7.11 - 6.99 (m, 2 H), 6.14 (s, 1 H), 4.54 - 4.44 (m, 1 H), 4.31 (d, j= 5.1 Hz, 3 H),4.13 (dd, J = 7.1, 10.3 Hz, 1 H), 3.83 (s, 3 H), 3.42 (q, J = 6.0 Hz, 2 H), 2.93 (br. s., 1 H), 2.71 - 2.56 (m, 4 H), 2.31 (br. s., 2 H), 2.19 (s, 3 H), 2.03 - 1.83 (m, 2 H), 1.64 (br. s., 1 H), 1.53 (d, ./.= 6.9 Hz, 3 H), 1.32 (d, J = 11.1 Hz, 1 H), 1.02 (d, J = 10.3 Hz, 1 H), 0.65 (d, J= 11.8 Hz, 1 H).
1003211 The compounds shown in the following table were prepared according to the general procedure outlined in this Example using the appropriate starting materials.
The structures of the compounds are shown in Figure 1.
Compound 1H mlz Name Number NMR
(R or S)-1-(1-(1-(2- NMR (400MHz, DMSO-d6) 8 11.58 (br. s., 1 H), hydroxy-2- 7.76 - 7.65 (m, 2 H), 7.58 (d, J = 7.8 Hz, 1 11), methylpropyl)piperidin- 7.10- 6.99 (m, 2 H), 6.14 (s, 1 H), 4.31 (d, J 4.9 4-yl)cthyl)-N-((4- Hz, 2 H), 4.14 (br. s., 1 H), 3.94 (s, 1 H), 3.83 (s, methoxy-6-methyl-2- 3 II), 3.56 (s, 2 H), 3.01 (d, J = 11.4 Hz, 1 H), 509 oxo-1,2-dihydropyridin- 2.73 - 2.64 (m, 1 H), 2.59 (s, 3 H), 2.19 (s, 3 H), 3-yl)methyl)-2-methyl- 2.16 -2.03 (m, 2 H), 1.52 (d, i = 6.9 Hz, 4 El), 11I-indole-3- 1.34 (br.
s., 2 H), 1.02 (d, J = 4.5 Hz, 7 H), 0.66 -carboxamide 0.58 (In, 1 Hi (R or S)-N-((4,6- (400MHz ,DMSO-d6) 6 11.59 (br. s., 1 H), 7.77 dimethy1-2-oxo-1,2- - 7.69 (m, 1 H), 7.60 (br. s., 2 H), 7.06 (br.
s., 2 dihydropyridin-3- H), 5.89 (s, 1 H), 4.31 (r, J = 5.7 Hz, 2 H), 4.20-yOmethyl)-1-(1-(1-(2- 4.09 (m, 1 H), 4.00 - 3.92 (m, 1 H), 3.58 -3.55 hydroxy-2- (m, 2 11), 3.19 - 3.10 (m, 1 H), 3.07 - 2.95 (m, methylpropyl)piperidin- H), 2.74 - 2.63 (m, 1 H), 2.59 (br. s., 3 H), 2.27 4-ypethyl)-2-methyl- (s, 3 H), 2.12 (s, 3 H), 1.89- 1.72 (m, 1 H), 1.54 1H-indole-3- (br. s., 4 H), 1.30- 1.14 (m, 2 H), 1.03 (br.
s., 6 carboxamide H), 0.84 - 0.57 (in, 2 H) 1003221 F. xarnple 31.
Synthesis of (R or S)-N-((4-methoxv-6-methyl-2-oxo-1,2-dihN (iron ridin-3-1,1)inethy 1)-2- ineth1.1-6-ohetwi-141-yetrahydro-2 Ii-pyran-4-yllethyll)-1H-intiole-3-rarboxanntk(C onwound 374), opH), r*, I )11,. H, : 1 0 IP C:
c, 0 WV 10 A 25 mL reaction tube was charged with a magnetic stir bar, phenyl boronic acid (72.6 mg, 0.596 mmol), K3PO4 (103 mg, 0.447 tumor), X-Phos pre-catalyst (Chloro(2-dicyclohexylphosphino-2',4',6'-triisopropy1-1,1'- bipheny1)[2-(2-am inoethyl)pheny1)] pallad ium(II)) (4.92 mg, 5.96 mop, and the vial was sealed. The vial was evacuatedlbackfilled with nitrogen (3x) before the addition of methyl 6-ehloro-2-methyl-1-(l -(tetrahydro-2H-pyran-4-ypethyl)-1H-indole-3-carboxylate (Compound 314) (100 mg, 0.298 mmol) as a solution in 1,4-dioxane (1 mL). The vial was then heated to 100 C
overnight with stirring. The vial was then allowed to cool to room temperature and the reaction concentrated in vacuo. The crude residue was purified via SiO2 chromatography (10g) using an eluent of ethyl acetate/hexanes (4:1) the title compound as a white solid (106 mg, 0.281 mmol, 94 A) yield).).
LCMS 514 (M+1)1.; III NMR 111 NMR (400MHz, DMSO-d6) 6 = 11.59 (s, 1 H), 7.98 -7.84 (m, 2H), 7.75- 7.67 (m,3 H), 7.47 (t, J = 7.8 Hz, 2 H),7.39 (d, J= 8.5 Hz, 1 H), 7.35 - 7.27 (in,!
H), 6.15 (s, 1 H), 4.35 (d, J= 4.9 Hz, 2 H).4.25 -4.12 (m, 1 H), 3.93 (d, J =
8.5 Hz, 1 H), 3.86 -3.77 (m, 3 H), 3.67 (d, J= 8.5 Hz, 1 H), 3.39 - 3.32 (m, 1 H), 3.10 - 3.00 (m, 1 H), 2.62 (s, 3 H), 2.21 (s, 3 H), 1.85 (d, J= 10.0 Hz, 1 H), 1.63 - 1.49 (in, 4 H), 1.45 - 1.33 (m, 1 H), 1.20 - 0.99 (m, 1 H), 0.66 (d, J= 12.0 Hz, 1 H).
[003231 Example 32. Synthesis of (R or SI-244-11-(3-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-v1)metlivIcarbamov1)-2-methyl-1H-indol-1-vflefird)piperidin-l-y1) acetic acid (Compound 364).
H LOH
''...(1-x P'`' ..,Nsr=-...C:õ N
. N
1 hi i I-IN 1 N i idik tiN N idik 0 0 4IF 0 0 lIF
To a round bottomed flask was charged with a magnetic stir bar was added (R or S)-ethy1-2-(4-(1-(3-((4-methoxy-6-methy1-2-oxo-1,2-dih ydropyridin-3-yl)methylcarbamoy 1)-2-methyl-LI-I-indo1-1-ypethyl)piperidin-1-y1)acetate (Compound 363) (69 mg, 0.132 mmol), THF
(1.5 mL), Me0H (1.5 mL), and water ( 0.75 mL). To this solution was added lithium hydroxide rnonohydrate (5.54 mg, 0.132 mmol) and the reaction stirred at room temperature for 1 h. The organics were removed under reduced pressure and the resulting aqueous solution purified via reverse phasc-HPLC (watcr/MeCN) 0-495% to afford the title compound (66 mg, 0.108 mmol, 82 % yield). LCMS 514 (M+1)' Ili NMR (400MHz ,DMSO-d6) 8 = 11.67 (s, 1 H), 9.65 (s, 1 H), 7.84 - 7.68 (m, 2 H),7.63 (d, J= 7.4 Hz, 1 H), 7.14 - 7.03 (m, 2 H), 6.18 (s, 1 H), 4.33 (d, J= 3.6 Hz, 2 H), 4.27 -4.15 (m, 1 H), 4.04 (br. s., 2 H), 3.85 (s, 3 H), 3.57 (s, 1 H), 3.35 - 3.23 (m, 1 H), 3.14 - 2.99 (m, 1 H), 2.86 - 2.74 (m, 1 H), 2.62 (s, 3 H), 2.21 (s, 3 H), 2.18 -2.08 (m, 1 H), 1.75 (s, 1 H), 1.60- 1.49 (m, 4 H), 1.46- 1.33(m, 1 H), 0.92 - 0.81 (m, 1 H).
[00324] Example 33. Synthesis of (R or S)-methyl 2-inetirvi-1-(1-(tetrahydro-211-thinpyran-4-vflethy0- 1 11-indole-3-ca rboxviate.
I10 '0 CI
,,,.........a ----10.
-0 lig 0 To a round bottomed flask charged with a magnetic stir bar was added (R or S)-methyl 2-methyl-1-(1-(tetrahydro-2H-thiopyran-4-ypethyl)-1H-indole-3-carboxylate (Step 6) (109 mg, 0.343 mmol) and DCM (5 inL). This solution was cooled to 0 C before addition of m-CPBA (154 mg, 0.687 mmol) and the reaction stirred at 0 "C for 30 minutes. The solution was then diluted with water and sat'd aqueous sodium thiosulfate solution and the layers separated. The aqueous was extracted with DCM and the combined organic layers were dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue was purified via silica gel chromatography (12 g) using ethyl acetatelhexanes (1:1) as eluent to afford the title compound (109 mg, 0.343 mmol, 95%). LCMS 350 (WO'. The title compound was used as an alternate starting material in Step 7 of Example 26 for the synthesis of other compounds of the invention.
[00325] Example 34. Synthesis of (R or S)-me1-11\ 1 2-metlryl-6-(rm ridi n-3-y1)- 141-ftetrahvdro-2H-Dvran-4-ynethyl)-1H-indole-3-carboxylate. This intermediate was used as an alternate starting material in Step 7 set forth in Example 26 for the synthesis of other compounds of the invention.
[00326] (R or S)-methyl 2-methyl-1-(1-(tetrahydro-2H-pyran-4-yl)ethyl)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y1)-1H-indole-3-carboxylate Ner0 Nr-N µ0 FICeHi 1)2 /
To a round bottomed flask was added Pd(OAc)2 (10.03 mg, 0.045 mmol), potassium acetate (219 mg, 2.233 mmol), 4,4,4',4',5,5,5',5'-octamethy1-2,2r-bi(1,3,2-dioxaborolane) (567 mg, 2.233 mmol), and 2-dicyclohexylphosphino-2' ,4' ,6' -triisopropylbiphenyl (XPhos) (85 mg, 0.179 mmol), and the vial was sealed. To this vessel was added (R or S)-methyl 6-chloro-2-methy1-1-(1 -(tetrahydro-2H-pyran-4-ypethyl)-1H-indole-3-carboxylate (Step 6) (500 mg, 1.489 mmol) dissolved in dioxane (3.4 mL) and the reaction evacuated/back-filled with N2 (3x) before heating to 100 C overnight. The reaction was then allowed to cool to rt and was diluted with Et0Ac. The reaction was filtered through diatomaceous earth and the filtrate concentrated to afford the title compound which was used in subsequent reactions without further purification.
LCMS 428 (M+1)4-.
[00327] (R or S)-methyl 2-methyl-6-(pyridin-3-y1)-1-(1-(tetrahydro-211-pyran-4-ypethyl)-1H-indole-3-carboxylate:
Br 8srjc--i PtiCla(cipp/J-CH2Cl2 Va. N
= ___________________ 40, vo.t.
To a re-sealable vial was added K2CO3 (206 mg, 1.488 mmol), PdC12(dpp1)-CH2C12 adduct (60.8 mg, 0.074 mmol), and the vial was sealed. This vial was evacuated/backfilled with N2 (3)() before addition of (R or S)-methyl 2-methy1-1-(1-(tetrahydro-2H-pyran-4-ypethyl)-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y1)-1H-indole-3-carboxylate (318 mg, 0.744 mmol) dissolved in 1,4-dioxane (4 mL), 3-bromopyridine (71.7 p.1, 0.744 mmol), and water (400 pi). The reaction was evacuatecl/backfilled with N2 (3x) before heating to 100 C. The solution was cooled to room temperature and diluted with Et0Ac. The solution was filtered and concentrated in vacuo. The crude residue was purified via silica gel chromatography (10 g, Et0Ac/hex (1:1)) to afford the title compound (101 mg, 0.267 mmol, 35.9 % yield). LCMS 379 (M+1)+.

1003281 The intermediates shown in the following table were prepared according to the general procedure outlined in this Example using the appropriate starting materials.
Name Structure µrei (R or S)-methyl 2-methy1-6-(pyrazin-2-y1)-1-(1-(tetrahydro-2H-pyran-4-yl)ethyl)- I H-indole-3-carbox.ylate 379 1 Ala .
(R or S)- methyl 2-m.ethyl.-1-(1-(tetrahydro-2H-pyran-4- -µo ypethyl)-6-(thiazol-4-y1)-1H-indole-3-carboxylate 0 = N

[003291 Example 35. Synthesis of (+)-N4(4,6-dimeth y1-2-oxo-1,2-dihydronvridin-yl)methyl)-1.-(1.-(4-fluarophen ) t vi)-2-methvl-lH-indole-3-earhoxamide (compound 1003301 ( )-1-(1-bromoethy1)-4-fluorabenzene.
Sr io Nfos._....1.146. 40 A mixture of 1-ethy1-4-fluorobenzene (0.248g, 2mmo1), NBS (0.35g, 2mmo1) and benzoyl peroxide (0.14g, 0.6mmo1) were dissolved in 20 mL CC14. The mixture was stirred at 80 C for 5 hours and then the reaction was conc. in vacuo and resulting oil was purified via silica gel chromatography (PE-Et0A.0 5: 1) to give the title compound (330 mg, 80%) as a yellow oil.
LCMS 202 (M F11).
[003311 The intermediates shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure ( )-1-(1-bromoethyl)-4-chloro benzene c * Br 218 Br ( )¨ I -(1-bromoethyl)-2-methoxybenzene 1101 2 0 6 ( )-1-bromo-3-(1-bromoethyl)benzene Br 110 264 N
( )-2-(1-bromoethyl)benzonitrile 210 Fir ( )-1-(1-bromocthy1)-3- 1110 m et hoxybenzene Br [00332] ( )-N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-(1-(4-fluorophenyl)ethyl)-2-methyl-1H-indole-3-earboxamide (Compound 166).
Flji * Br F
)1117 HN N
N-((4,6-di methy 1-2-oxo-1,2-dihydropyridin-3-yl)methy I)-2-methy I-1H-indol e-3-carboxamide (309 mg, 1.0mmo1) was dissolved in 4 mL DMF, NaU (80mg, 2.0mmo1) was added, the mixture was stirred at room temperature for 30 min, and then 1-(1-bromoethyl)-4-fluorobenzene (0.404mg, 2.0mmol) was added. The mixture was stirred at room temperature overnight and was directly purified by reverse phase HPLC (A:CH3CN,B:water+0.1% MCI. A:B=35:65 150*25mm) to afford the title compound as a white solid(11 mg, yield 3%). LCMS
432 (M +
114) 1H NMR (400 MHz, CDCI3) 6 7.78 (d,J=8Hz, 1H), 7.33 (m, 1H), 7.14 (s, 1H), 6.96 (m, 2H), 6.92 (in, 1H), 6.85-6.92 (m, 4H), 5.85 (s, 1H), 5.72 (m, 1H), 4.56 (s, 2H), 2.64 (s, 3H), 2.37 (s, 3H), 2.11 (s, 3H), 1.85(d, .1=7.2 Hz, 31).
[00333] Example 36. Synthesis of ( )-1-(1-(4-ehloronhenyflethyD-N-((2-hydroxy-4,6-dimethylpyridin-3-vpmethyl)-2-methyl-1H-indole-3-earboxamide (Compound 170).
1003341 Step 1: Ethyl 2-methyl- I H-indo1e-3-carboxylate.

_NH2 A}La"..s-s"
_______________ 1.=
Cu2O, 0S2CO3 0 To a mixed solution of dimethyl sulfoxide (100 triL) and water (34 mL), 2-iodobenzenamine (50 g, 228 mmol), ethyl 3-oxobutanoate (35.6 g, 274 mmol), copper(I) oxide (3.3 g, 22.8 mmol) and cesium carbonate (75 g, 228 mmol) was added. The mixture was stirred at 100 C
for 16 hours under nitrogen gas atmosphere. The reaction mixture was filtered through a pad of celite. The filtrate was diluted with water and extracted with ethyl acetate. The organic phase was concentrated in vacuo, and then the residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 5:1) to afford the title compound as a light yellow solid (26.6 g, 57.5%). LCMS 204 (M + H)+.
[003351 The intermediates shown in the following table were prepared according to the general procedure outlined above for ethyl 2-methyl-IFI-indole-3-carboxylate using the appropriate starting materials and modifications.
Name Structure ink ethyl 6-chloro-2-methyl-1H- 239 indole-3-carboxylate tert-butyl 2-methyl- 1 H- 254 indole-3-carboxylate o 0 (M + Na-) ethyl 2-methy1-6-(methylsulfony1)- I H-indole- oAlla 282 3-carboxylate Vir SO2Me [00336] Step 2: ( )-ethyll-(1-(4-chlorophenyi)ethyl)-2-methyl4H-indole-3-carboxylate:
CI
Br CI

To a solution of ethyl 2-methyl-1H-indole-3-carboxylate (250 mg, 1.23 mmol) in anhydrous DMF (2 inL) was added NaH (60 A) in mineral oil, 74 mg, 1.85 mmol) at room temperature under N2. The reaction was stirred at 50-60 C for 30 min. Then the reaction was cooled to 0 C
and a solution of 1-(1-bromoethyl)-4-chlorobenzene (from Example 35; 400 mg, 1.85 mmol) in DMF (1 mL) was added drop-wise. The reaction was stirred at room temperature overnight.
Then the mixture was diluted with water and extracted with Et0Ac. The organic extract was combined, dried over Na2SO4 and concentrated in vacuo. The residue was purified by silica gel chromatography using an eluent of petroleum ether/Et0Ac (60:1) to afford the title compound as a yellow oil. (210 mg, yield 50.1%) NMR (Methanol-d4, 400 MHz) 8 8.07-8.04 (m, 1H), 7.33-7.29 (m, 211), 7.16-7.15 (m, 2H), 7.14-7.08 (m, 1H), 7.07-6.96 (m, 2H), 5.97 (q, Jj = 7.2 Hz, ../2 = 14.4 Hz, 111), 4.37 (d, J = 7.2 Hz, 2H), 2.76 (m, 3H), 1.95 (d, J =
6.8 Hz, 3H), 1.43 (d,./
= 14.0 Hz, 3H).
1003371 Step 3: ( )-1-(1-(4-ch1orophenyl)ethyl)-2-methyl-1H-indo1e-3-carboxylic acid:
*1 KOH
H =
0 =
To a mixture of ethyl 1-(1-(4-chlorophenyl)ethyl)-2-methyl-IH-indole-3-carboxylate (210 mg, 0.61 mmol) in Me0H/F120 (6 mL/2 mL) was added KOH (340 mg, 6.1 mmol) at room temperature. The reaction was rcfluxed overnight. Then the mixture was adjusted to Ph = 4 with 1 N HC1 and extracted with Et0Ac (3x). The combined organic extract were combined and concentrated in vacuo to afford the title compound as a yellow solid (200 mg, yield 105 %) which was used without further purification.
1003381 Step 4: ( )-1-(1-(4-chlorophenyl)ethyl)-N-((2-hydroxy-44-dimethylpyridio-3-y1)methyl)-2-methyl-1H-indole-3-carboxamide (Compound 170):
ci * CI FIN
HO I

0 NriN
HN N

[003391 To a solution of 1-(1-(4-chlorophenypethyl)-2-methy1-1H-indole-3-carboxylic acid (200 mg, 0.64 mmol) in anhydrous DCM (5 rnL) was added HOBt (130 mg, 0.96 mmol), EDCI
(184 mg, 0.96 mmol) and Et3N (194 mg, 1.92 mmol) at room temperature under N2.
The reaction was stirred for 30 min and then 3-(aminomethyl)-4,6-dimethylpyridin-2-ol (107 mg, 0.7 mmol) was added. The reaction was stirred at room temperature overnight. The mixture was diluted with water and adjusted to pH 7 and extracted with DCM. The crude product was purified by silica gel chromatography (Eluent: DCM:Me0H=30: 1) to afford the title compound a yellow solid. (120 mg, yield 41.9 %) 'H NMR (DMSO-d6, 400 M Hz) 5 11.6 (s, 1H), 7.74-7.68 (m, 2H), 7.39 (dd,.// = 2.0 Hz, J2 = 6.8 Hz, 2II), 7.16 (d, .1= 8.4 Hz, 211), 7.06-6.93 (m., 311), 5.92 (t, J.= 7.2 Hz, 2H), 4.33 (d, J.= 5.6 Hz, 211), 2.59 (s, 3H), 2.26 (s, 3H), 2.11 (s, 31-1), 1.87 (dõ I... 7.2 Hz, 3H); ESI-MS: tn/z 447.8 [M -4- Hr.
1003401 Example 37. S%nthesis of ( 1-1-(1-bromoethvi)-3-methribenzene.
40 PBr) ______________ 11-OH Br Phosphorus tribromide (4.28 g, 15.9 mmol) was added drop-wise to a stirred neat solution of 1-(m-tolyl)ethanol (0.9 g, 6.6 mmol) at 0 C. Alter being stirred to room temperature over 12 h, the reaction was carefully quenched with sat'd saturated aqueous NaHCO3 solution and the mixture was extracted with Et0A.c. The organic extract was washed with water, dried with MgSO4, and conc. in mow to afford the title compound (1.2 g, 91%) as a colorless oil, that was used directly in the next step without further purification. LCMS 200 (M
1003411 The intermediates shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
Name Structure miz ( )-1-(1-bromoethyl)-4- ID Me 200 methylbenzene õBr Me ( )-1-(1-bromoethyl)-2-methyl benzene Br ( )-1-(1-bromoethyl)-4-)4 264 (methylsulfonyl)benzene _______________________________ Br 0 (1)-1-(1-bromoethyl)-3-chlorobenzene CI
Br Br 0 ( )-1-(1-bromoethy4-3- µ,00 (methylsulfonyl)benzene hydroxyethyl)benzonitrile N
OH
1003421 These intermediates were used in place of ( )-1-(1-bromoethyl)-4-fluorobenzene in Example 35 or 1-(1-bromoethyl)-4-chlorobenzene in Example 36, as appropriate, to make other compounds of the invention.
[003431 Example 38. Synthesis of ( )-1-(pyriditte-4-ybethvi 4-meiln ibenzenesuifonate.
TsCI
_op 0 *LC

OH N
To a solution of 1-(pyridine-4-ypethanol (400 mg, 3.25 mmol) in THF (10 mL) was added sodium hydride (301 mg, 12.54 mmol) at 0 C. The mixture was stirred at 0 C
for 30 min. 4-methylbenzenc-l-sulfonyl chloride (744 mg, 3.90 mmol) was added and the mixture was stirred at room temperature for 4 hr. Water (10mL) was added and THF was removed under reduced pressure. The residue was extracted with ethyl acetate (20 mLx3). The organic layer was washed with brine and dried over sodium sulfate. The solvent was removed under vacuum and the residue was purified by column chromatography on silica gel (eluted: petroleum ether/ethyl acetate = 2/1) to give 1-(pyridine-4-yl)ethyl 4-methylbenzenesulfonate (0.56 g, 62.2%). LCMS
278 (M H1).
[003441 The intermediates shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
Name Structure miz ( )-1-(pyridine-3-yl)ethyl ft `crit.7) 278 4-methylbenzenesulfonate 0 ----- -( )-1-(pyridine-2-ypethyl 4 278 4-methylbenzenesulfonate ( )-1-(1-m et hyl -IEI- 0 I ,0 ,..
pyrazol-4-ypethyl 4- 0 6, "0 ...- N., 281 methylbenzenesulfonate ¨rµr ( )-1-(2-methoxypyridin- 4 ito 4-yl)ethyl 4- 6:'-0 ...- N. 308 methylbenzenesulforiate ( )-1-methoxypropan-2-y1 ...4.
0¨= o_L¨."% 169 methanesulfonate 0 ( )-1 -ethoxypropan-2-y14-methylbenzenesulfonate Si O' i (+)-1 -(5-methoxypyridin- 1 41 4' 3-yl)ethyl 4- / .cykriC1 308 0 , I
methylbenzenesulfonate= t4 1-(2-methoxypyrimidin-4- 4 p ypethyl 4- .
i 1 6) ..-c y0 -- 309 methylbenzeriesulforiate 1 -0=,- il 3-methoxybutan-2-y1 4- 0 411 p 259 methyl benzenesulfonate it-crii ...

(. )-4-((tert- o butyldimethylsilyl)oxy)but an-2-y1 methanesulfonate 1 1003451 These intermediates were used in place of ( )-1-(1-bromoethyl)-4-fluorobenzene in Example 35 or 1-(1-bromoethyl)-4-chlorobenzene in Example 36, as appropriate, to make other compounds of the invention.
[003461 Example 39. Other Alkyl Carboxvlate Intermediates. The following alkyl carboxylate intermediates were synthesized in an analogous manner to that set forth in Step 2 of Example 36, using an appropriate starting material and reactant.

Name Structure miz =
( )-ethyl 2-methy1-1-(1-(p-tolypethyl)-1H-indole-3- ; 321 carboxylate 0\
(74-ethy1 2-m ethy1-1-(1-(o-tolypethyl)-1H-indole-3- 321 carboxylate =, ( )-ethyl 2-methyl- 1-( I -(m-tolybethyD-1H-indole-3- N

carboxylate 0 aliks ( )-ethyl 2-m ethy 1-1-(1-(4-(methylsulfonypphenypethyl)- õa.õ, N 386 111-indole-3-carboxyl ate tpi ( )-ethyl 1-(1-(3-chlorophenyflethyl)-2-methyl- 4 343 1H-indole-3-carboxylate () Name Structure mlz ( )-ethyl 1-(1 -(2-methoxyphenyl)ethyl)-2-methyl- N 338 1 H-indole-3-carboxylate Br _________ ( )-ethyl 1 -(1 -(3-bromophenypethy 1)-2-methyl- N 387 1H-indole-3-carboxylate 1 Am -( )-ethyl 2-methyl- 1 -(1-(3- \)" ft A
(methylsulfonyl)phenypethyl)- N .,3' I Alk \ 386 1H-indole-3-carboxylate ( )-ethyl 2-methyl- 1 -( 1 -(pyridine-4-ypethyl)-1H-indolc-3-carboxylate ,0 ( -ethyl 2-methyl- 1 -(1 -(pyridine-3-ypethyl)-1H-indole- 1 309 3-carboxy la te ( )-ethyl 2-methyl-1 -(1 - \r. ..
(pyridine-2-ypethyl)-1H-indolc- N 309 ha-L.
3-carboxylate ethyl 2-methyl- 1 43-methyloxetan-3-yl)methyl )- 1 H- 288 indole-3-carboxylate ( )-ethyl 2-methyl-1 -(1 -(1 -N
methyl-1 H-pyrazol-4-y 1)ethyl 312 1H-indole-3-carboxylate ,\1`L.:
y ( )-ethyl 1 -(1 -(2-methoxypyridin-4-yDethyl)-2-methyl-1H-indole- 339 3-carboxylate Name Structure mk j).- __________________________________________________ ( )-ethyl 5-fluoro-1-(1- \r-methoxypropan-2-y1)-2-methyl- 294 111-i ndole-3-carboxyl ate ( )-ethyl 6-fluoro-1-(1- )--µ
methoxypropan-2-y1)-2-methyl- r 294 1H-i ndole-3-carboxyl ate -...... A\
1114.r F
( )-ethyl 1-(1-methoxypropan-2- )--' y1)-2-methy1-1H-indole-3- )rN 276 carboxylate ( )-ethyl 1-(1-(3- ,.s.
cyanophenyl)ethyl)-2-methyl-1H- Tr" Chi 333 indole-3-carboxylate 0 -( )-ethyl 1-(sec-buty1)-6-chloro- \ri NI
2-methyl-I H-indole-3- 1 295 carboxylate -......,0 411, lir/ ci NC
( )-ethyl 1-(1 -(2- .
cyanophenypethyl)-2-methy1-1H-indole-3-carboxylate \ i o _ (1)-1(sec-buty1)-2-methy1- I H- o Y"--pyrrolo[2,3-b]pyridine-3- . .4 il 289 carboxyl ate ''=== ''' N
N
\ /

( )-tert-butyl 1-(1-\1_ j methoxypropan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-o 1 305 carboxylate o \o ( )-tert-butyl 1-(1-(3-\ra methoxyphenyl)ethyl)-2-methyl- \ /

11-1-pyrrolo[2,3-b]pyridine-3-carboxylate Name I Structure mk ( )-tert-butyl 14.1-(2-....... - - ". 6 methoxypyridin-4-yl)ethyl)-2- \ /

methyl-1H-pyrro lo [2,3-b]pyridi ne-3-carboxyl ate \ /

( )-tert-butyl 1-(1-ethoxypropan- \--J
2-y1)-2-methy1-1H-pyrrolo[2,3- 4 319 b]pyridine-3-earboxylate _________________________________________ -----------------------\ /
o GO-tea-butyl 1-(1-cyanoethyl)-2- ).......-71:-N
N
methyl-1 H-pyrrolo[2,3- 286 b]pyridine-3-earboxylate st 1 C;
o \o ( )-tert-butyl 14 145-methoxypyridi n-3-yl)ethyl)-2- s\r......6 \ pi 368 methyl-1H-pyrro lo [2,3-b]pyridine-3-carboxylate ...., __ ( )-tert-butyl 1-(1-(2-methoxypyrimidin-4-ypethyl)-2- 369 methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate \ /
- i 0 ( Heft-butyl 2-methyl- I -(1-(--) morpholi nopropan-2-yI)-1H - )......7 pyrrolo [2,3-b]pyridine-3- 360 N
carboxylate ( )-tert-butyl 1-(141H-\I: ik benzo[d]imidazol-1-yl)propan-2- , 391 y1)-2-methyl-1H-pyrrolo [2,3- N
b]pyridine-3-carboxylate >r \ /
N
'I
( )-tert-butyl 1-(1-(3-cyanophenypethyl)-2-methyl-IH- i 362 pyrro lo [2,3-b]pyridine-3- r0 N
carboxylate >
. n Name I Structure to¨ mk tert-butyl 1-(3-methoxybutan-2-y1)-2-methyl-1H-pyrrolo [2,3- 319 ,,)4, 0 js N).,..N
b]pyridine-3-carboxylate ( )-tert-butyl 1-(1-(3-_________________________ L

c0N,i2 carbamoylphenypethyl)-2-) 380 methy1-1H-pyrro I o [2,3- . i b]pyridi ne-3-carboxyl ate \ /

( )-tert-butyl 2-methy1-1.-(1-(2- fil \rf 0 oxopyridin-1(2H)-yl)propan-2-y1)-1H-pyrrolo[2,3-b]pyridine-3-carbo xyl ate >nor -µ) ( )-tert-butyl 2-methyl- I -(1-Nj (m ethy Isulfonyl)propan-2-y1)-1.1:1- r pyrrolo[2,3-b) pyridine-3- 01.(1.61N_N 353 carboxy late ( )-tert-butyl 2-methyl- I -(1-\)--i -V.
(pyridine-2-yloxy)propan-2-y1)- cõry...:6 368 1H-pyrrolo[2,3-b]pyridine-3- i ....N
carbox.ylate )-- . 0 "
ethyl 1-(3-methoxybutan-2-yI)-2- 4--methy1-6-(methylsulfony1)-1H- N

indole-3-carboxylate .......õ,o *
SO2Me N _______ ( )-tert-butyl 1-(1-cyanopropan- ') __ /
2-y1)-2-methyl- I H-pyrrolo[2,3- 300 rõo,X.ct.
bilpyridine-3-carboxylate / N
( )-tert-butyl 1-(4-((tert- , ____ . --- 01"83 ? .. ' butyl dimethylsilypoxy)butan-2- N.--N:.

y1)-2-methyl-IH-pyrrolo [2,3- . . o. -1.,õ" i, . ir \
b]pyridi ne-3-carboxyl ate : 0 ..- , tert-butyl 2-methyl-1-(piperi din-1.
, -ylsulfony1)-1H-pyrrolo [2,3- \ NP 0 tr- 380 b]pyridine-3-carboxylate 0 ';'" = , ,Il ='-"N
=::, ;', ( )-tert-butyl 1-(2-((tert- .---/
0ISS __ butyldimethylsilyl)oxy)propy1)-2- - .ri \
)1. .., 405 methyl-1H-pyrrolo [2,3- .õ.o,,,.,.J., b]pyridinc-3-carboxylate I

Name Structure rn/z.
, pme ( )-ethyl 1-(3-methoxypentan-2-\----P1 \
y1)-2-methy1-1H-indole-3- & s>--- 304 carboxylate ( )-tert-butyl I -( I -methoxypropan-2-y1)-2-methy1-6- , N 0-'171 (trifluoromethyl)-1H-pyrrolo[2,3- )r.c 1 -- N
b]pyridine-3-carboxylate II / cF3 -- ---------(0-ethyl 6-(4-(tcrt-butoxycarbonyl)piperazin-l-y1)-141-methoxypropan-2-y1)-2- -,,n I " ; 461 methyl-1H-pyrrolo[2,3- , ¨ ta 0 1 / NsTh b]pyridine-3-carboxylate Ls/ Boc [00347] Example 40. Other Carboxylic Acid Intermediates. The following carboxylic acid intermediates were synthesized in an analogous manner to that set forth in Step 3 of Example 36, using an appropriate starting material (e.g., one of the alkyl carboxylate intermediates set forth in the previous Example).
Name Structure rn/z.
WI
Me ( )-2-methy1-1-(1-(p-tolypethyl)-1H-indole-3- :6 293 carboxylic acid HO
\ /
( )-2-methy1-1-(1-(o- .
tolyl)cthyl)-1H-indolc-3- \õir.N 293 carboxylic acid nolf2,,,b (- )-2-methy1-1-(1-(m- 1#it tolyl)ethyl)-1H-indole-3- N.,...Ni 293 carboxylic acid I
HO
I 11) =
( )-2-methy1-1-(1-(4- z, * so (methylsulfonyl)phenyl)ethyl) 0.1õlb 358 - I H-indole-3-carboxylic acid H
I \ /
C

Name Structure rniz, 1 f 1 chlorophenypethyl)-2-methyl- ,rrNibN 315 1H-indole-3-carboxylic acid HO
\ i i methoxyphenyl)ethyl.)-2- .

methyl.-1H-indole-3-...1k) carboxylic acid HO
\ I

Br ( )-1. -(l -(3-bromophenyl)ethyl)-2- lik methyl-111-indok-3- N

carboxylic acid HO *
( )-2-methy1-1-(143- _=__=;) ,,S' (methylsulfonyl)phenyfl N ethyl) 358 -1H-indole-3-carboxylic acid HO

( )-2-methy1-1-(1-(pyridine-N.,_õõN
4-ypethyl)-1H-indole-3- 11 >=) 281 carboxylic acid t o .
( )-2-methy1-1-(1-(pyridine- i N
3-34)ethyl)-1H-indole-3- 281 carboxylic acid Ho.N_Dt4 o \ i ( )-2-m ethy1-1-(14pyridine-2-ypeth y1)-1. H-indole-3-11., - 281 carboxylic acid HO...1( i....\)) 0 .................................................. --1 1-(1-(1,4-dioxan-2-yOethyl)-N
2-methyl-1H-indole-3- 290 I
carboxylic acid HO 4 1-(1-(14tert- )..<70c butoxycarbonyl)piperidin-3- N 387 yl)ethyl)-2-methy1-1H-indolc- i õin Ai 3-carboxylic acid 0 1-Er Name Structure rniz, 1 ) --0(F
di fluorocyclohexy1)133yri d)- N F 322 2-methyl- I 11 -indole-3- HO I all carboxylic acid o 141111"-2-Methyl-1-((3-methyl ox e tan- rNco ..1.11õ, .be 3-yl)methyl)-1H-indole-3- 260 carboxylic acid HO \ /
t -/
( )-2-methyl-1-(1-(1-methyl-1H-pyrazol-4-ypcthyl)-1H- i N 283 indole-3-carboxylic acid Ho 4 .:, ( )-2-methyl-1-(1-(1-methyl- -A
\)õ..... 1( 2-oxo-1,2-dihydropyridin-4-\--N 310 ypethyl)-1H-indole-3-HOstf,X6 carboxylic acid C
03oc 1-(1-(tert-butoxycarbonyl)piperid in-4-y1)-2-methy1-1H-indole-3- I
Ho am carboxylic acid 0 mir ck..., 2-methy1-1-(quinolin-5-y1)- N -.--.1 1H-indole-3-carboxylic acid 1 1-cycl openty1-2-m ethy I-1H- ..iN
P

indole-3-carboxylic acid HO I AIN

( )-5-fluoro-1-(1-`, =N
methoxypropan-2-yI)-2-methyl-1H-indole-3- H0I ,11 4 266 carboxylic acid o \
F ¨4 ( )-6-fluoro-1-(I- -, .2 ¨
meth oxyp ropan -2-y1)-2-methyl-1H-indole-3-carboxylic acid 0 F
\ p ¨
( )- 1 - ( 1 -methoxypropan-2-\¨N
y1)-2-methyl-1H-indo le-3- 248 carboxylic acid / i\
\.:..,..:õ/

Name Structure rniz 1 cyanophenyl)ethyl)-2-methyl- CONH2 HO 323 ...i.YbN
1 H-indole-3-carboxylic acid ,-, 2-methyl-1-(6- * ) methylqui nol in-5-y 0-1H- N 317 ';---i ndole-3-carboxylic acid 1 ( )-1-(sec-buty1)-6-chloro-2- \ri methyl-IH-indole-3- 267 Ht., , ¨
carboxylic acid I
( )-2-methy1-1-(1-(1- )-011.
(methylsulfonyl)azetidin-3- N 8 ypethyl)-1H-indole-3- HO I
carboxylic acid o 030c ( )-1-(1-(2-(4-(tert-butox.ycarbonyl)piperazin- I - ,, yppyridine-4-yDethyl)-2- ),¨ N'. _if 465 methyl-1H-indole-3- N
I
carboxylic acid HO ilk 0 11411r*
( )- 1. 41.42- \r-clj cyanophenyl)ethy4-2-methyl- N..-N 305 111-indole-3-carboxylic acid HO il 1-(2,5-dimethylpheny1)-2- .
methy1-1H-indole-3- N 280 carboxylic acid I
..,.
1-(2,5-dimethylpheny1)-2-methy1-1H-in dole-3- NNI...-N 280 carboxylic acid /
HOI, 4 N.._ 2-methyl-1-(quinolin-6-y1)- . 1 1H-indole-3-carboxylic acid \t¨N 303 Name Structure rniz 0¨
1-(3-methoxybutan-2-y1)-2-methy1-6-(methylsulfony1)- 340 1H-indole-3-carboxylic acid Ho so,me NrC
(R or S)-2-methyl-1-(1-(tetrahydro-2H-pyran-4-ypethyl)-1H-pyrrolo[2,3- H0 b]pyridinc-3-carboxylic acid /\
(S)-2-methy1-1-(1-HO /
phenylethyl)-1H-pyrrolo[2,3- 281 b]pyridine-3-carboxylic acid , ph*
1-(3-methoxypentan-2-y1)-2-methyl-1H-indole-3-carboxylic acid 110, \.
( )-6-(4-(tert-butoxycarbonyl)piperazin-1-y1)-1 -(1-methoxypropan-2-y1)-2-methyl-11-1-pyrrolo[2,3- N'Th bipyridine-3-carboxylic acid [003481 Examvle 41. Other Comnounds of the Invention Produced from Carboxylic Acid Intermediates. The following compounds were synthesized in an analogous manner to that set forth in Step 4 of Example 36, using an appropriate starting material (e.g., one of the carboxylic acid intermediates set forth in the previous example). Structures of these compounds are set forth in Figure 1.
Compound Name 114 NMR miz ( )-N-((4,6-dimethy1-2-(400 MHz, CDC13) ö 11.39 (s, 1H), 7.76 (d, = 8.0 Hz, 1H), 7.30 (t, J = 6.4 Hz, 1H), 7.02-oxo-1,2-dihydropyridin-3-172 6.87 (m, 7H), 5.83 (s, 1H), 5.72 (q, J = 6.4 A,"
ypmethyl)-2-methyl-1-(1 (p-tolypethyl)-1H-indole-3--Hz, 1H), 4.59-4.50 (m, 2H), 2.63 (s, 3H), 2.37 carboxamide '1 (s, 3H), 2.23 (s, 3H), 2.10 (s, 3H), 1.83 (d, J =
7.2 Hz, 3H).

Compound Name NMR ink (400 MHz, CDC13) 8 1.80 (s, 3 H) 1.88 (d, J=7.03 Hz, 3 H) 2.17 (s, 3 H) 2.44 (s, 3 H) ( )-N-((4,6-dimethy1-2-2.65 (s, 3 H) 4.61 (d, 1=6.02 Hz, 2 H) 5.78 (q, oxo-1,2-dihydropyridin-3-174 J=7.11 Hz, 1 H) 5.91 (s, 1 H) 6.91 - 6.97 (m, 4.)8 yOmethyl)-2-methyl-1-(1-1 H) 6.98 - 7.05 (m, 2 H) 7.08 (d, 1=7.53 Hz.
(o-tolypethyl)-1H-indole-3-1 H) 7.20 - 7.25 (m, 1 H) 7.28 - 7.40 (m, carboxamide H) 7.61 (d, J=7.78 Hz, 1 H) 7.81 (d, 1=8.78 Hz, 1 H) (400 MHz, Methanol-d4) 8 7.83 (d, .1=7.21-1z ( )-N-04,6-dimethy1-2-, 111), 7.42 (s, 11-1), 7.24 (s, 1H), 7.14-7.18 oxo-1,2- dihydropyrid in-3-190 (m, 2 H), 6.92-6.99 (m, 5 H), 5.92 (s, 1H), 418 yOmethyl)-2-methyl-1-(1-5.78 (m, 1 H), 4.62 (s, 2 H), 2.70 (s, 3 H), (m-tolypethyl)-1H-indole-2.43 (s, 3H) , 2.16 (s, 3H), 1.99 (s, 3H) , 1.89 3-carboxamide (s, 1H), 1.88 (d, J=7.2 Hz, 3H) ( )-N-((4,6-dimethyl-2- (400MHz, CDC13) 8 ppm 2.00 (d, J=7.06 Hz, oxo-1,2-dihydropyridin-3- 3H) 2.20 (s, 3H) 2.46 (s, 3H) 2.72 (s, 3H) 179 yOmethyl)-2-methyl-1-(1- 3.04 (s, 3H) 4.57 - 4.71 (m, 2H) 5.85 (q, (4- 1=7.20 Hz, 1H) 5.95 (s, 1 H) 6.89 (d, 1=8.16 (methylsulfonyl)phenypeth Hz, 1H) 6.98 (t, 1=7.61 Hz, 1H) 7.02 - 7.08 y1)-1H-indole-3- (m, 1H) 7.32 (d, J=8.16 Hz, 2H) 7.47 (t, carboxamide 1=5.84 Hz, 1H) 7.87 (d, 1=8.38 Hz, 3H) ( )-1-(1-(3- (400 MHz, CDC13) 5 7.71-7.79 (d, 1F1), 7.31-chlorophenybethyl)-N- 7.77 (t, 1H), 7.11-7.16 (m, 3H), 6.94-6.99 (m, 181 ((4,6-dimethy1-2-oxo-1,2- I F1), 6.87-6.92 (m, 311), 5.84 (s, 111), 5.67-dihydropyridin-3- 5.72 (m, 1H), 4.53-4.56 (t, 211), 2.64(s, 311), yl)methyl)-2-methyl-1H- 2.37 (s, 3H), 2.11 (s, 3H), 2.57 (s, 3H), 1.83-indole-3-carboxarnide 1.88 (d, 3H) ( )-N44,6-dimethy1-2- (400 MHz, CDC13) 5 7.72 (d, .1=7.2 Hz,! H), oxo-1,2-dihydropyridin-3- 7.39 (d, J=7.2 Hz,1H), 7.28 (s, 1H)õ 7.16-186 yOmethyl)-1-(1-(2- 7.19 (m, 2H), 6.83-6.92 (m, 3H), 6.72 (d, methoxyphenypethyl)-2- J=8.4 Hz, 1H), 5.88 (d, J=11.2 Hz, 2H), 4.55 methyl-1H-indole-3- (s, 2H), 3.51 (s, 3H), 2.73 (s, 3H), 2.36 (s, carboxamide 3H), 2.10(s, 3H), 1.73(d, .1=6 Hz, 3H) ( )-1-0 -(3-(400 MHz, CDCI3) 8 7.47-7.38 (m, 3H), 7.14-bromophenyl)ethyl)-N-7.10 (t, 1H), 6.99-6.94 (m, 4H), 5.92 (s, 1 H), ((4,6-dimethy1-2-oxo-1,2-5.76-5.74 (dd, 1H), 4.64-4.61 (dd, 211), 493 dihydropyridin-3-2.70(s, 3 H), 2.43 (s, 3 H), 2.14 (s, 3 H), 1.90-yl)methyl)-2-methy1-1H-1.88 (t 3H) indole-3-carboxamide Compound Name 'H. NMR Ink ( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(3- 492 (methylsulfonyl)phenypeth y1)-1H-indole-3-carboxamide N 4-methoxy-6 (CDC13, 400MHz) 5 8.55 (s, 2H), 7.87-7.85 -()--0 (d, J=7.6Hz, 1 H), 7.61 (s, 1H), 7.05-6.97 (m methy1-2-oxo-1,2-4H), 6.88-6.86 (d , J=8.0Hz , 1H. 5.95 (s, 387 dihydropyridin-3-111), 5.78-5.76 (d, J=7.2Hz, 111), 4.69-4.68 431 yl)methyl)-2-methy1-1-(1-(d, J-5.2Hz , 2H) 3.92 (s, 3H), 3.16-3.05 (in, (pyridine-4-ypethyl)- I H-1H), 2.73 (s, 3H) , 2.21 (s, 3H) , 1.97-1.95 (d, 1ndole-3-carboxamide J=7.2Hz, 3H) (CD30D, 400 MHz) 6 8.86-8.84 (d, J =5.6, ( )-N((4-methoxy-6- 1H), 8.74 (s, 1H), 8.37-8.35 (d, J =7.2, I H), methy1-2-oxo-1,2- 8.09-8.06 (m, J =14.4, 111.), 7.82-7.80 (d, J
392 dihydropyridin-3- -8.0, 1H), 7.21-7.18 (t, .1 -7.2, 1H), 7.11-7.07 Ali yOmethyl)-2-methyl-1-(1- (t, J =7.6, 1H), 7.03-7.01 (m, 2H), 6.29-6.24 (pyridine-3-y1)ethy1)-1H- (q, J -7.2, 1H), 4.67 (s, 2H), 4.17 (s, 3H), indolc-3-carboxamide 2.77 (s, 311), 2.58 (s, 3H,), 2.13-2.12 (d, J
=7.2,3K) (CD30D, 400 MHz) 6 8.56-8.54 (d, J = 4.4 ( )-N-((4-methoxy-6- Hz, 1H), 7.79-7.75 (t, J = 7.2 Hz, 1H), 7.72-methy1-2-oxo-1,2- 7.70 (d, J = 8.0 Hz, 1H), 7.36-7.33 (t, J = 7.2 393 dihydropyridin-3- Hz, 1H), 7.19-7.17 (d, J =8.0 Hz, 1H), 7.05-yOmethyl)-2-methyl-1-(1- 7.03 (d, J =8.0 Hz, 1H), 6.97-6.95 (m, 2H), (pyridine-2-yflethyl)-1H- 6.42 (s, 1H), 6.03-5.98 (q, J = 7.2 Hz, 1H), indole-3-carboxamide 4.55 (s, 2H), 3.98 (s, 3H), 2.65 (s. 3H), 2.36 (s, 3H), 1.99-1.97 (d, J = 7.2 Hz, 3H) ( (450101:mizilzciDj),1\47S704-(W7665= (1m1:603 17.H2,(8d.(5191(d..., 4.1;
264 .)-N-((4,6-dimethy1-2-oxo-1,2-dibydropyridin-3- 7.1 Hz, 1 H), 7.09 ¨ 7.05 (m, 2 H), 7.00 (t, J = 7.4 Hz, yOmethyl)-2-methyl-1-(1- ID, 6.95 (t, J = 7.7 11z, III), 6.00 (ch./ ¨ 7 .1114 1 415 (pyridine-2-yl)ethyl)-1H- H), 5.89 (s, 1 H), 4.33 (d, J = 5.2 Hz, 2 H), 2.62 (s, 3 indole-3-carboxamide H), 2.27 (s, 3 H), 2.12 (s, 3 H), 1.90 (d, 1 =
7.1 Hz, 3 11) (400 MHz, CDC13) 6 1.63 (d, J=7.2, 3H), 2.14 (s, 3H), 2.70 (s, 3H), 3.18-3.14 (m, 2H), 3.60-1-(1-(1,4-dioxan-2-3.53 (m, 114), 3.69-3.67 (m. 1H), 3.81-3.75 ypethyp-N-((4-methoxy-6-(m, 1H), 3.18-3.14 (in, 1H), 3.86 (s, III), 3.89 348 methy1-2-oxo-1,2-(s, 3H), 4.30-4.25 (m, 1H), 4.43-4.36 (m, 1H), 440 dihydropyridin-3-4.67-4.65 (m, 21), 5.91 (s, 1H), 7.05-7.01 (m, yOmethyl.)-2-methy1-1H-1H), 7.12-7.08 (m, 1H), 7.47-7.44 (m, 1H), indole-3-carboxamide 7.59-7.56 (m., 1H), 7.84-7.83 (m, 1H), 12.85 (s, 1H) Compound Name 'H. NMR ink (400MHz, METHANOL-d4) 8= 7.75 (d, J=8.0 Hz, 1H), 7.57 (d, J=7.5 Hz, 1H), 7.15 N-04-methoxy-6-methyl-2- (quin, J=6.5 Hz, 2H), 6.31 (s, 1H), 4.62 (s, oxo-1,2-dihydropyridin-3- 1H), 4.56 (s, 2H), 4.38 ¨ 4.28 (m, 1H), 3.97 yl)methyl)-2-methy1-1-(1- (s, 3H), 3.57 (d, J=11.0 Hz, 1H), 3.23 (d, 437 (piperidin-3-yl)ethyl)-1H- J=12.5 Hz, 1H), 2.85 ¨2.74 (m, 2H), 2.64 (s, indole-3-carboxamide 3H), 2.35 (s, 3H), 1.72 (s, 1H), 1.69 ¨ 1.62 (m, 3H), 1.47 ¨ 1.30 (m, 1H), 1.15 ¨ 1.04 (m, 2H) (CDC13, 400MHz) ö 12.63-12.64 (d, .1 =3.2 Hz, 1H), 7.84(s, 1H), 7.49 (s, 1H), 7.42-7.40 ( _)-1-(1-(4,4-(d, j =9.2 Hz, 1H), 7.06-7.00 (m, 2H), 5.90-difluorocyclohexypethyl)-5.89 (d, .1 ¨3.6 Hz 1H), 4.66-4.62 (t, J =14 304 N-04-methoxy-6-methy1-2-Hz, 211), 4.11-4.08 (m, 1H), 3.88-3.87 (d, J 427 oxo-1,2-dihydropyridin-3-=3.6 Hz, 3H), 2.99-2.76 (m, 3H), 2.36 (s,1H), yl.)methyl)-2-m ethy1-1H- 2.25 ( . s, 311), 2.17-2.16 (d, J =3.2 Hz, 2H), I*ndo1e-3-carbox. ide 2.08-2.05 (m, 2H), 1.84-1.70 (m, 2H), 1.61 (s, 1H), 1.51-1.47 (m, 211) (400MHz, d6-DMS0) 8 11.60 (br. S. 1 H), N-04,6-dimethy1-2-oxo- 7.79 7.74 (m, 1 11), 7.73 ¨ 7.67 (m, 1 H), 1,2-dihydropyridin-3- 7.52 7.47 (m, 1 H), 7.18 7.11 (m, 1 H), yl)methyl)-2-methy1-1-((3- 7.10 7.05 (m, 1 H), 5.89 (s, 1 H), 4.51 (d, J 394 methyloxetan-3-ypmethyl)- = 5.8 Hz, 2 H), 4.33 (d, J = 5.6 Hz, 2 14), 4.30 1H-indolc-3-carboxamide (s, 2 H), 4.10 (d, .1 = 6.0 Hz, 2 H), 2.54 (s, 3 H), 2.26 (s, 3 H), 2.12 (s, 3K), 1.27 (s, 311) ( )-N-((4,6-dimethyll::2-oxo-1,2-dihydropyridin-3- (400MHz, DMSO-d6) 6 11.69 (s, 1 H), 7.74 ¨ 7.64 (m, 745 yl)methyl)-1-methyl-1-(1- 2 H), 757 (s, 1 H), 7.28 7.23 (in, 2 H), 7.04 6.96 (m, 2 II), 5.93 (s, 1 II), 5.79 (q, J= 7.3 Liz, 1 II), 4.35¨ 418 (1-methy1-1H-pyrazol-4- 4.31 (m, 2 H), 3.76 (s, 3 H), 2.64 Is. 3 H), 2.28 (s, 3 H), ypethyl)-1H-indole-3- 2.13 (s,3 H), 1.79 (d, J= 7.1 Hz, 3 H) carboxamide ( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-222 yOmethyl)-2-methy1-1-(1-(1-methy1-2-oxo-1,2-dihydropyridin-4-ypethyl)-1H-indole-3-carboxarnide tert-butyl4-(3-(((4- (400 MHz, CDC13) 8 11.9 (s, 1H), 7.85-methoxy-6-methy1-2-oxo- 7.82(d, 1H),7.51-7.41 (m, 2H),7.10-7.01 (m, 331 1,2-dihydropyridin-3- 2 H), 5.90 (s, 1H), 4.64 (s ,2H), 4.37(s, 1 H), 509 Amethypcarbamoy1)-2- 3.89 (s, 3 H), 2.84 (s, 2 11) , 2.75 (s, 3 H), methyl-1H-indo1-1- 2.49 (s, 2 Ft) .2.17 (s, 3H) .1.79-1.76 (d, yl)piperidine-1-carboxy1ate 21-0,1.50 (s, 9H) Compound Name 'H NMR m/z (CDCI3, 400 MHz) 8 8.97 (dd, Jj - 1.6 Hz, J2 = 4.4 Hz, lip, 8.31 (d, J = 8.4 Hz, 1H), 8.01 N-04-methoxy-6-methyl-2- (d, J = 8.0 Hz, 1H), 7.88 (t, J = 8.8 Hz, 1H), oxo-1,2-dihydropyridin-3- 7.76 (s, 1H), 7.56 (d, ./= 7.2 Hz, 1H), 7.40 (d, yl)methyl)-2-methyl-1- J= 8.4 Hz, 1H), 7.31-7.28 (m, 1H), 7.15 (t, J 453 (quinolin-5-y1)-1.F1-indole- = 7.2 Hz, 1H), 7.03 (t, J = 7.6 Hz, 1H), 6.71 3-carboxamide (d, J = 8.0 Hz, 1H), 5.97 (s, 1H), 4.80-4.68 (m, 2H), 3.93 (s, 3H), 2.44 (s, 3H), 2.29 (s, 3H) 1-cyclopentyl-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3- 394 yl)methyl)-2-methy1-1H-indole-3-carboxamide ( )-5-fluoro-N-((4-methoxy-6-methy1-2-oxo- (400 MHz, CD30D) 8 7.59-7.55 (m, 1H), 1,2-dihydropyridin-3- 7.42-7.39 (m, 1H), 6.95-6.90 (m, 2H), 4.57 (s, yl)m.ethyl)-1-(1- 2H), 4.12 (s, 3H), 3.99-3.94 (m, 1H), 3.72- 416 methoxypropan-2-y1)-2- 3.65 (m, 1H), 3.19 (s, 3H), 2.64(s, 3H), 2.54 (s, 3H), 1.59-1..57(d, 3H) carboxamide ( )-6-fluoro-N-04-methoxy-6-methy1-2-oxo- (400 MHz, CD30D) 6 7.70-7.66 (m, 1H), 2 1,2-dihydropyridin-3- 7.36-7.33 (m, 1H), 6.94-6.89 (m, 2H), 4.56 (s, yl)methyl)-1-(1- 2H), 4.11 (s, 3H), 3.97-3.92 (m, 1H), 3.71- 416 methoxypropan-2-y1)-2- 3.67 (m, 1H), 3.20 (s, 3H), 2.62(s, 3H), 2.53 methyl-1H-indole-3- (s, 3H), 1.58-1.56(d, 3H) carboxamide ( )-N4(4-m.ethoxy-6-methy1-2-oxo-1,2- (4(K) MHz, CD30D) 8 7.69 (d, 1=7.2 Hz,1H), 218 dihydropyridin-3- 7.53 (d, .11=7.6 Hz,1H), 7.12 (m, 2H), 6.26 yOmethyl)-1-(1- (s,IF1), 4.80(m,1H), 4.52 (s,211), 3.99 (m, methoxypropan-2-y1)-2- 4H),3.75 (m, 1H),3.20 (s, 3H), 2.62 (s, 3H), methyl-1H-indole-3- 2.31 (s, 311), 1.59 (d, J=7.2 Hz, 3H) carboxamide ( )-N-((4,6-dimethyl.-2-(400 MHz, CD30D) 8 7.74 (m, 1H), 7.57 (d, oxo-1,2-dihydropyridin.-3- -J=7.6 Hz, 1H), 7.15 (m, 2H), 6.14 (s,1H), 1 83 yl)methyl)-1. -(1-4.86 (m, 1H), 4.55(s, 2H), 4.02 (m, 1H),3.77 382 methoxypropan-2-y1)-2-(m., 1H),3.22 (s, 3H), 2.65 (s, 3H), 2.43 (s, methy1-1H-indole-3-311), 2.26 (s, 311), 1.62 (d, J=7.2 Hz, 311) carboxamide Compound Name 'H NMR m/z ( )-1-(1-(3-(400 MHz, Methanol-d4) 6 7.84 (d, cyanophenyl)cthy1)-N-(4,6-.1-811z,1H), 7.66 (d, j=8.4 Hz, 2H), 7.33-7.37 175 dimethy1-2-oxo- 1 2-(m, 111), 7.04 (m, 1 H), 6.88-6.96 (m, 2H), dihydropyridin-3-y1)-2-, 5.90 (s,111),5.81-5.82 (m, 1 H), 4.57-4.63 (m, 457 methyl-1H-indole-3-2 H), 3.54 (s, 1 H), 2.99(s, 3H), 2.70(s, 3H), carboxamidc 1.45(s, 3H), 1.22(s, 3H), 1.96(d, J=7.2 Hz, 3H) N-((4-methoxy-6-methy1-2-(CD30D, 400 M Hi) 8 9.27 (s, 11-1), 8.48 (m oxo-1,2-dihydropyridin-3-114), 8.35 (s, III), 8.13 (s, 1F1), 8.01 (s, 11-1), yl)methyl)-2-methy1-1-(6-390 7.95 (d, J=7.06 Hz,1H), 7.29 (s, 1H), 7.15 (s, methyl quinolin-5-y1)-1H-111), 7.01 (s, 111), 6.68 (s, 1H), 4.67 (s, 2H), indole-3-carboxa.mide 467 4.16 (s, 3H), 2.57 (s, 3H), 2.33 (s, 3H), 2.21 (s, 3H) ( )-1-(sec-butyl)-6-chloro- (400 MHz, CD30D): 67.71 (d, J=8.8, 1H), 385 N-04-methoxy-6-methyl-2- 7.63 (s, 1H), 7.15 (d, 1=8.8, 1H), 7.00 (s, 1H), oxo-1,2-dihydropyridin-3- 4.60 (s, 3H), 4.15(s, 3H), 2.67(s,3H), 2.57 (s, 416 yl)methyl)-2-methy1-1H- 3H), 2.15-2.25 (m, 1H), 1.95-2.01(m, 1H), indole-3-carboxamide 1.62(d, J=7.2, 3H), 0.73(t, J=7.6, 3H) ( )-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-(CDC13, 400MHz) 6 12.52 (s, 1 H), 7.86-7.03 354 yl)methyl)-2-methyl-1-(1-(m, 511), 5.92 (s, 1 H), 4.71-4.62 (m, 3H), 4.16-4.12 (m, 111), 3.90 (s, 3 H), 3.79-3.32 487 (1- (methylsulfonyl)azetidin-3- (m, 4H), 2.80 (s, 6 If), 2.18 (s, 3 H), 1.28 (d, J ypethyl)-1H-indole-3-8EIz, 3H) carboxamide (-.0-N-04-methoxy-6-NMR (4(10 MHz, CDC13) c 8.10-8.11(d, J
methyl-2-oxo-1,2-=
5.6 Hz, 11-1), 7.72-7.75 (d, J= 8.0 Hz, 1H), dihydropyridin-3-6.98-7.11 (m, 3E1), 6.57-6.61 (m, 2H), 6.30 (s, 353 yl)methyl)-2-methyl-1-(1-1H), 5.87-5.92 (m, 1H), 4.55 (s, 1H), 3.96 (s, (2-(piperazin-1-y1) 515 3H), 3.66-3.69 (m, 4H), 3.22-3.24 (d, J = 2.8 pyridine-4-yl)ethyl)-1H-Hz, 3H), 1.96-2.99 (d, = 9.6 Hz, 114), 2.63 indole-3-carboxamidc (s, 3H), 2.33 (s, 3H), 1.94-1.96 (d, J = 7.6 Hz, 3H), 1.30-1.34 (m, 3H) N((4-methoxy-6-methy1-2-(CDC13, 400 MHz) 5 7.91 (d, J = 7.2 Hz, oxo-1,2-dihydropyridin-3-1H), 7.59 (s, 1H), 7.12-7.04 (m, 3H), 6.89 (d, 339 yl)methyl)-2-methyl-1-J= 7.6 Hz, 1H)' 6.59 (d, J= 8.0 Hz, 1H), 6.48 (1,2,3,4-tetrahydroquinolin-(d, J = 7.6 Hz, 1H), 5.95 (s, 1H), 4.75-4.64 457 5-y1)-1H-indole-3-(m, 211), 3.91 (s, 3H), 3.27 (t, J - 6.0 Hz, 21-1), carboxamide 2.50 (s, 3I1) 2.27 (s, 3FI), 2.10-1.98 (m, 21-1), 1.77-1.63 (m, 211) Compound Name 'H NMR m/z (400 MHz, CDC13) 8 ppm 2.02 (d, 1=6.62 Hz, (1)-1-(1 -(2-3 H) 2.19 (br. S., 3 H) 2.44 (br. S., 3 H) 2.77 cyanophenyl)ethyl)-N-(hr. S., 3 H) 4.62 (br. S., 2 H) 5.92 (hr. S., 1 178 ((4,6-dimethy1-2-oxo-1,2-H) 5.96 - 6.05 (m, 1 H) 6.92 - 7.12 (m, 3 H) 439 dihydropyridin-3-7.33 - 7.50 (m, 3 H) 7.54 - 7.62 (m, 1 H) yl)methyl)-2-methy1-1H-7.66 (d, J=6.84 Hz 1 H) 7.86 (d, 1=7.50 Hz, indole-3-carboxamide ' 1 H) (400 MHz, CDC13) 6 10.86 (s, 111), 7.91 (d, N-(4,6-dimethy1-2-oxo-= 8.0, 1H), 7.46 (s, 111), 7.29-7.19 (m, 2H), 1,2-dihydropyridin-3-191 7.12 (d, .1=7.2, 111), 7.06 (d, I = 7.2, 11-1), yl)methy,5-6.96 (s, 1H), 6.78 (d, J = 8.0, 1H), 5.91 (s, dimethy1pheny1)-2-methy1-1H), 4.69-4.57 (m, 2H), 2.45 (s, 3H), 2.44(s, 1H-indole-3-carboxamide 3H), 2.35 (s, 3H), 2.22 (s, 3H), 1.85 (s, 3H) N-((4,6-dim.ethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-(2,3- 414 dimethylpheny1)-2-methyl-1H-indole-3-carboxamide (Me0D, 400 M Hz) 8 7.88 (d, 1=7.94Hz, N-04-methoxy-6-methy1-2-1H), 7.56-7.51 (m, 21), 7.26 (t, 1=7.5Hz, oxo-1,2-dihydropyridin-3-1H), 7.18 (t, 1=7.61Hz, 1H), 7.16 (s, 1H), 391 yOmethyl)-2-methyl-1-(6-6.76 (d, J=7.94Hz, 1H), 4.63 (s, 2H), 4.14 (s, 471 3H), 3.51 (t, 1=5.29Hz, 2H), 2.56 (s, 3H), tetrahydroquinolin-5-y1)-2.33 (s 3H), 2.31-2.20 (m 1H) 2.18-2.14 (m 1H-indole-3-carboxamide ' " ' 111), 2.02-1.99 (m, 2H), 1.89 (m, 3H) N-04-methoxy-6-methyl-2- (400 MHz, METHANOL-d4) 8 ppm 7.82 (d, oxo-1,2-dihydropyridin-3- .1=7.94 Hz, 1 H) 7.60 (d, .1=8.38 Hz, 1 H) 7.37 386 yl)methyl)-2-methyl-1- - 7.48 (m, 2 H) 7.13 - 7.28 (m, 2 H) 6.97 (1,2,3,4-tetrahydroquinolin- 7.07 (m, 2 H) 4.62 (s, 2 H) 4.15 (s, 3 11) 3.57 6-y1)-1H-indole-3- - 3.66 (m, 2 H) 3.06 (t, 1=6.28 Hz, 2 H) 2.56 carboxamide (s, 3 H) 2.49 (s, 3 H) 2.16 - 2.29 (m, 2 H) N-04-methoxy-6-methyl-2- (400 MHz, METHANOL-4) 8 1.03 (d, oxo-1.2-dihydropyridin-3- .7=6.62 Hz, 6 H) 2.32 (s, 4 H) 2.70 (s, 3 H) yOmethyl)-2-methyl-1-(3- 2.96 (dõ1=7.06 Hz, 2 H) 3.95 (s, 3 H) 4.52 (s, 410 methylbutanoy1)-1H- 2 H) 6.28 (s, 1 H) 7.20 - 7.30 (m, 2 1-0 7.66 indole-3-carboxamide (d, .1=6.84 Hz, 1 H) 7.91 (d, 1=7.72 Hz, 1 H) N-04-methoxy-6-methy1-2-(400 MHz, CD30D): 5 7.78-7.76 (d, 111), oxo-1,2-dihydropyridin-3-7.68-7.66 (d, iii), 7.33-7.26 (m, 211), 6.97 (s, 372 yl)methyl)-2-methy1-1-1H), 4.58 (s, 211), 4.12 (s, 311(), 3.95-3.94 (m, 438 (tetrahydro-2H-pyran-4-2H), 3.63-3.57 (m,1H), 3.54-3.52 (m, 2H), carbony11-1H-1ndole-3-2.69 (s, 3H), 2.55 (s, 31-1), 1.88-1.82(m, 4H) 1 carboxamide Compound Name NMR m/z (400 MHz, CDC13) 6 11.14 (s, 111), 8.16 (s, ( )-(4,6-dimethy1-2-oxo-11-1), 8.12 (d, f = 7.6 Hz, 1H), 7.32 (t, .1= 5.6 1,2-dihydropyridin-3-Hz, 1H), 7.25-7.01 (m, 2H), 7.05-7.01 (m, 201 yl)methyl 1-("),3-dihydro-2H), 6.76 (d, J = 7.6 Hz, 1H), 5.86 (s, 1H), 428 1H-inden-1-y1)-2-methyl-4.52-4.50 (m, 2H), 3.13 (bs, 1H), 3.05-2.97 1H-pyrrolo[2,3-b]pyridine-(m, 1H), 2.66 (bs, 1H), 2.73 (s, 3H), 2.29 (bs, 3-carboxylate 2H), 2.13 (s, 3H) ( )-1-(see-buty1)-N-04,6- (400 MHz, CDC13) dimethy1-2-oxo-1,2- a 8.116-8.132 (d, 1H), 8.011-8.035 (d, 11-1), 185 dihydropyridin-3- 7.037-7.057(t, 14), 6.056 (s, 1H), 4.462 (d, yl)methyl)-2-methyl-IH- 211), 6.30 (s, 111), 2.63(s, 3H), 2.356 (s,3H), pyrrolo[2,3-b]pyridine-3- 2.189(s, 3H), 1.880-1.933 (m, 1H), 1.587-earboxamide 1.605 (d, 2H), 1.226 (s, 2H), 0.658 (t, 3H) (400 MHz, CDC13) 6 11.03 (s, 1H), 8.15 (dd, J1 = 4.8 Hz, J2 = 1.6 Hz, 1H), 8.06 (dd, ii =
( )-(4,6-dimethy1-2-oxo- 8.0 Hz, J2 = 1.6 Hz, 1H), 7.32 (t, J = 7.0 Hz, 1,2-dihydropyridin-3- 1H), 6.97 (dd, J, = 8.0 Hz, J2 =4.8 Hz, 1H), Amethyl 2-methyl-1- 5.86 (s, 1H), 5.47-5.39 (m, 1H), 4.52 (d, ./ =

(tetrahydrofuran-3-y1)-1H- 7.0 Hz, 2H), 4.39-4.32 (m, 1H), 4.17 (dd, .././ =
PYITo1o[2,3-b]pyridine-3- 9.2 Hz, J2 = 6.8 Flz, 1H), 3.98 (t, J = 8.8 Hz, earboxylate 1H), 3.91-3.86 (q, J = 7.6 Hz, II1), 2.76 (s, 3H), 2.59-2.50 (m, 1H), 2.36 (s, 3H), 2.34-2.28 (m, 1H), 2.25 (s, 3H) ( )-N-((4-methoxy-6- (400 MHz, CDC13) 6 13.23 (s, 1H), 8.16-8.17 methy1-2-oxo-1,2- (m, 1H), 8.11-8.13 (in, 1H), 7.57-7.60 (t, J =
204 dihydropyridin-3- 5.2Hz, 1H), 6.93-6.96 (m, 1H), 5.92 (s, 1H), yOmethyl)-1-(1- 4.82-4.83 (d, J= 2.4 Hz, 1H), 4.65-4.66 (d, J

methowropan-2-y1)-2- = 6.4 Hz, 2H), 3.89 (s, 3H), 3.81-3.85 (rn, methyl-1H-pyrrolo[2,3- 1H), 3.22 (s, 3H), 2.79 (s, 3H), 2.17 (s, 3H), b]pyridine-3-carboxamide 1.64-1.66 (d, J= 8.0 Hz, 3H) ( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-206 yOmethyl)-1-(1-(3-methoxyphenyl)ethyl)-2-methyl-IH-pyrrolo[2,3-b]pyridine-3-earboxamide (.. )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yOmethyl)-1-(1-(2-methoxypyridin-4- 446 yl)ethyl)-2-methy1-114-pyrrolo[2,3-b]pyridine-3-carboxarnide Compound Name 1H NMR Ink 1-(chroman-4-y1)-N-((4,6-dimethy1-2-oxo-1,2-209 dihydropyridin-3-yl)methyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (4)-1-(1-cyclopropylethyl)-N4(4,6-dimethy1-2-oxo-211 1,2-dihydropyri di n-3-Amethyl )-2-m ethyl-1H-pyrrolo[2,3-b]pyridine-3-carboxamide ( )-1-(1-ethoxypropan-2-y1)-N-((4-methoxy-6- (400 MHz, CDC13) 8 8.173-8.189 (m, 1H), 8.13-8.153 (m, 1H), 7.563(s, 1H), 6.977-methy1-2-0x0-1,2-212 7.008 (m, 1H), 5.938 (s, 1H), 4.652-4.667 (d, dihydropyridin-3-yl)methyl )-2-m ethy1-1H- 21.1), 4.177(s, 1H), 3.309-3.454 (m, 2H), 3.94-3.98(m, 1H), 2.806 (s , 3H), 2.212 (s, 3H), pyrrolo[2,3-b]pyridine-3-1.665-1.682 (d, 3H), 1.044 (t, 3H) carboxamide ( )-1-(1-cyanoethyl)-N44-((4 (400 MHz, Methanol-d4) 8 8.28-8.26 (dd. .//
= 4.8 Hz, .12 = 1.2 Hz, 1H), 8.12-8.09 (dd, J-1=
methoxy-6-methy1-2-oxo-214 1,2-dihydropyridin-3- 4.8 Hz, .12 = 1.2 Hz, 111), 7.21-7.18 (dd, J1=
8.0 Hz, .1.2 = 4.8 Hz, 1H), 6.26 (s, 1H), 6.16- 379 yl)methyl)-2-methyl-1H-6.11 (q. J= 7.2 Hz, 1H), 4.50 (s, 2H), 3.93 (s, pyrrolo[2,3-b]pyridine-3-carboxamide 3H), 2.76 (s, 3H), 2.31 (s, 3H), 1.88 (d, J=
7.2 Hz, 3H) ( )-N44,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yOmethyl)-1-(1-(5-methoxypyridin-3- 446 yl)ethyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide =
(400 MHz, CDC1::) 8 12.74-12.79 (m, 1H), 8.16-8.17 (d, J= 4.0 ( )-1-(1-methoxypropan-2- Hz, 1H), 8.08-8.10 (d, J= 4.0 Hz, 1H), 7.45-yl)-2-methyl-N46-methyl- 7.48 (t, J= 5.6 Hz, 1H), 6.93-6.96 (m, 1H), 216 2-oxo-4-propy1-1,2- 5.96 (s, 1H), 4.81 (s, 1H), 4.61-4.62 (d, J=
dihydropyridin-3- 5.6 Hz, 2H), 4.17-4.22 (t, J= 8.4Hz, 1H), 411 yl)methyl)-1H-pyrrolo[2,3- 3.80-3.84 (m, 1H), 3.22 (s, 3H), 2.78 (s, 3H), b]pyridine-3-carboxamide 2.71-2.75 (t, J= 8.0 Hz, 2H), 2.18 (s, 3H), 1.64-1.65 (d, J= 6.8 Hz, 5H), 0.98-1.02 (t, J
= 7.6 Hz, 311) Compound Name 111. NMR miz ( )-N-((4,6-dimethy1-2-oxo-11-dihydropyridin-3- (CDC13, 400 MHz) .5 11.82 (s, 1H), 8.24-8.15 , 220 (m, 2171), 7.25-7.18 (m, 5H), 7.04 (dd, f, =
4.8 yl)methyl)-2-methy1-1-(1-Hz, .12 = 8.0 Hz, 1H), 6.19 (s, 1H), 5.93 (s, 429 phenylpropy1)-1H-111), 4.58 (s, 211), 2.64-2.50 (m, 5H), 2.42 (s, pyrrolo[2,3-b]pyridine-3-311), 2.17 (s, 3E1), 0.81 (t, J = 7.6 Hz, 311) earboxamide (CDC13, 400 MHz) 5 12.03 (s, 1171), 8.23 (dd, ( )-1-(1-(1H-pyrazol-1- Jj 1.2 Hz, J2 = 4.4 Hz, 11i), 8.12 (dd, J./ =
yl)propan-2-y1)-N-((4,6- 1.2 Hz, J2 = 7.6 Hz, 1H), 7.43 (d, J= 1.6 Hz, dimethy1-2-oxo-1,2- 1H), 7.31 (t, J= 6.0 Hz, 1H), 8.12 (q, .// =
4.8 dihydropyridin-3- ilz, .12 = 8.0 Hz, 111), 6.58 (d, J = 2.0 Hz, 1H), 419 yOmethyl)-2-methyl-IH- 5.93 (d, 10.8 Hz, 111), 5.91 (d, J=
2.0 Hz, pyrrolo[2,3-b]pyridine-3- 1H), 5.35 (q, .// = 10.0 Hz, .12 = 13.2 Hz, 1H), earboxamide 4.97 (s, 111), 4.62-4.54 (m, 3H), 2.41 (s, 6H), 2.19 (s, 3H), 1.74 (d, = 6.8 Hz, 3H) ( )-N4(4-methoxy-6-methy1-2-oxo-1,2-(400 MHz, Me0D-d4) 6 8.56-78.54 (d, I H), dihydropyridin-3-8.34-8.32 (d, 111), 8.26-8.25 (d, 1H), 7.37-232 yOmethyl)-1-(1-(2-7.34 (t, 1H), 7.05-7.02 (t, 1H.), 6.28-6.26 (d. 463 rnethoxypyr1m1din-4-1H), 4.63(s, 2H), 4.16(s, 3H), 3.9(s, 311), yl)ethyl)-2-methy1-111-2.71(s,311), 2.59(s, 311), 2.15-2.13(d,3171) pyrrolo[2,3-b]pyridine-3-carboxarnide ( )-N((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yOmethyl)-1-(1-(2-methoxypyrimidin-4- 447 ypethyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (400 MHz, CDC13) 6 ( )-N-((4-ethoxy-6-methyl- 12.5 (s, 111), 8.11-8.18 (m, 2H), 7.60 (s, 1H), 2-oxo-1,2-dihydropyridin- 6.95-6.98 (rn, 1H), 5.90 (s, 1H), 5.96 (s, 1H), 235 3-yl)methyl)-1-(1- 4.83 (s, 111), 4.10-4.21 (m, 311), 3.82-3.83 (m, methoxypropan-2-y1)-2- 1H), 3.23 (s, 3H), 3.79 (s, 3H), 2.15 (s, 3H), methyl-1H-pyrrolo[2,3- 1.65-1.66 (d, J = 6.8 Hz, 6H), 1.44-1.47 (t, .1 b)pyridine-3-carboxamide = 7.2 Hz, 3H).
( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-237 yl)methyl)-2-methyl-1-(1-morphoIinopropan-2-y1)-1H-pyrrolo[2,3-b]pyridine-3-earboxamide Compound Name 'H NMR Ink ( )-1-(1-(1H-benzo[d]imidazo1-1-yl)propan-2-y1)-N-04,6-238 dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (0-1-043- (400 MHz, CD30D) 6 8.18-8.12 (m,11-1), cyanophenyl)ethyl)-N-((4-8.10 (d, J=6.4 Hz,' H), 7.61-7.57 (m, 1H), methoxy-6-methy1-2-oxo-239 7.45- 7.44 (m, 2F1), 7.16-7A3 (m, III), 6.31 1,2-dihydropyridin-3-(d, J-:8.4 Hz, 111), 6.25 (s, 1H), 4.50 (s, 21-1), 4'56 yl)methyl)-2-met- 1H-3.91 (s, 311). 2.50 (s, 3H), 2.30 (m, 311), 2.05 pyrrolo[2,3-b]pyridine-3-(d, J=7.2 Hz, 3H) carboxamide N-04,6-dimethy1-2-oxo-(400 MHz, CD30D): 8 8.67-8.65 (d, 1H), 1,2-dihydropyridin-3-8.45-8.44 (d, 1H), 7.59-7.55(m, 1H), 6.70 (s, 241 yl)methyl)-1-(3-1H), 4.79 (s, 1H), 4.61 (s, 2H), 4.07 (s, 1H), 397 methoxybutan-2-y1)-2-3.32 (s, 3H), 2.75(s, 3H), 2.56 (s, 3H), 2.42(s, methy1-1H-pyrrol 3H), 1.68-1.66(d, 3H), 1.16-1.15(d, 31).
blpyridine-3-carboxamide 04-141 -(3-carbamoylphenypethyl)-N- (400 MHz, CD30D-d4) 8 8.31 (br, 211), 7.78-((4-methoxy-6-methy1-2- 7.79 (br, 1H), 7.77 (s, 1H), 7.46-7.33 (m, 3H), oxo-1,2-dihydropyridin-3- 6.97 (s, 1H), 6.50-6.48 (d, J = 6.8 MHz, 1H), 474 yOmethyl)-2-methyl- 1H- 4.60 (s, 2H), 4.41 (s, 3H), 2.56 (s, 3H), 2.53 pyrrolo[2.3-b]pyridine-3- (s, 3H), 2.11-2.10 (d, J= 7.2 MHz, 3H) carboxamide N-((4,6-dimethy1-2-oxo-(400 MHz, CD30D) 8 8.30 (m, /If), 8.25 (m, 1,2-dihydropyridin-3-111), 7.29 (m 1 HI 6.85(s, 114) 4.77 (m 244 yl)methyl)-2-methy1-1- " " ' ' 1H),4.62 (s, 2 IT), 4.13 (m, 2f1), 3.65 (m, 2 395 (tetrahydro-2H-pyran-4-y1)-FT), 2.99 (m,211), 2.78 (s, 3H), 2.60 (s, 3I1), 1H-pyrrolo[,3-b]pyridine-2.46 (s, 3H), 1.77 (m, 2H) 3-carboxamide (CDC13, 400 MHz) 6 11.18 (s, 1H), 8.24 (dd, Jj = 1.2 Hz, J2 = 4.8 Hz, 1H), 8A3 (dd Jj =
( )-N44,6-dimethy1-2-1.2 Hz, J2 = 9.2 Hz, 1H), 7.30 (t, J = 6.0 Hz, oxo-1,2-dihydropyridin-3-1H), 7.20-7.15 (m, 1H), 7.06 (dd, Ji = 4.8 Hz, ypmethyl)-/-methyl-1-(1-249 J2 = 8.0 Hz, 1H), 6.50-6.43 (m, 2H), 5.93 (s, (2-oxopyridin-1(2H)- 446 1H), 5.70-5.66 (in, 1H), 5.11 (s, 1H), 4.80 (t, ).l)propan-2-34)-1H-J = 12.4 Hz, 1H), 4.69 (dd, Ji = 4.4 Hz, pyrrolo[2,3-b]pyridine-3-12.8 Hz, Iff), 4.53 (dd, J,= 3.2 Hz, J2 = 5.6 carboxamide H7, 214), 2.50 (s, 3H), 240 (s, 3f1), 2.20 (s, 3F1), 1.74 (d, J= 7.2 Hz, 31-I) WO 2013/120104 PCT1tJS2013/025639 Compound Name 'H NMR Ink ( )-N-((4,6-dimethy1-2- (400 MHz, CD30D): 8.31-8.32 (d, J = 6.0 oxo-1,2-dihydropyridin-3- Hz, 3H), 8.18-8.20(d, J - 8.0 Hz, 3H), 7.26-250 yl)methyl)-2-methyl-1-(1- 7.29 (t, 1H), 6.99 (s, 111), 5.20-5.37 (br, 1H), (methylsulfonyl)propan-2- 4.58 (s, 2H), 4.13 (s, 3H), 3.67-3.71 (d, 1H), y1)-1H-pyrrolo[2,3- 3.1-3.22 (hr, 1H). 2.78 (s, 3H), 2.55 (s, 3H), bipyridine-3-carboxamide 2.54 (s, 3H),1.77-1.79 (d, J= 6.8 Hz, 3H).
(400MHz, CDC13) 8 ppm 1.80 (d, J-6.84 Hz, ( )-N-((4-methoxy-6-3 H) 2.21 (s, 3 H) 2.73 (s, 3 If) 3.90 (s, 3 H) methy1-2-oxo-1,2-4.66 (d, J=5.51 Hz, 2 H) 4.82 4.90 (m, 1 H) dihydropyridin-3-251 5.11 (d, J=9.04 Hz, 2 H) 5.93 (s, 1 H) 6.57 (d, yl)methyl)-2-methy1-1(1- 462 J=8.38 Hz, 1 H) 6.79 - 6.86 (m, 1 H) 7.01 (pyridine-2-yloxy)propan-(dd..1-7.94, 4.85 Hz, 1 li) 7.45 - 7.52 (m, 1 2-y1)-1H-pyrrolo[2,3-H) 7.58 (br. S., 1 H) 8.07 - 8.17 (m, 2 H) 8.18 b]pyridine-3-carboxamide - 8.23 (m, 1 H) 11.63 (br. S., 1 H) (CDC13, 400 MHz) 8 11.04 (s, 1H), 8.18 (d, J
( )-N-((6-cyclopropy1-4- = 4.0 Hz, 1H), 8.10 (d, J = 8.0 Hz, 1H), 7.28 methoxy-2-oxo-1,2- 4, I = 6.0 Hz, 1H), 7.02 (dd, Ji = 4.8 Hz, J2 =
260 dihydropyridin-3- 7.6 Hz, 1H), 5.77 (s, 1H), 4.82 (s, 1H), 4.57 yOmethyl)-1-(1- (d, = 6.0 Hz, 211), 4.19 (t, = 8.8 Hz, 1H), 425 methoxypropan-2-y1)-2- 3.83 (ci, .11 = 5.6 Hz, .12 - 9.6 Hz, 1F1), 3.22 (s, methy1-1H-pyrrolo[2.3- 311), 2.77 (s, 3H), 2.41 (s, 311), 1.74-1.68 (m, b]pyridine-3-carboxamide 111), 1.65 (d, J = 7.2 Hz, 3H), 0.90-0.76 (m, _411) (400 MHz, CDC13): 8 0.97-0.99 (d, J=6.8 Hz, N(4,6-dimethy1-2-oxo- 3H), 8 1.19-1.21(d, 1=6.0 Hz, 3H),2.16(s, 1,2-dihydropyridin-3- 3H), 2.41(s, 3H), 2.73(s, 3H), 2.99(s, 3H) 322 yOmethy1)-1-(3- , 3.04 (s, 311), 3.859 (brs, 111), 4.47(brs, methoxybutan-2-y1)-2- 1H), 4.59-4.61 (d, J=6.0 Hz , 2H), 5.95(s, methyl-6-(methylsulfony1)- 111), 7.41-7.44 (t, J=5.6 Hz,1H), 7.52-7.54 1H-indole-3-carboxamide (d, 1=4.8 Hz , 1H), 7.97-7.99(d, 1=8.4 Hz, 1H), 8.117(s, 1H) (400 MHz, CDC13) 6 1.20-1.24 (d, J=6.4 Hz, N((4-methoxy-6-methy1-2- 3H), 8 1.59-1.69(d, J=6.4 Hz, 3H),2.13(s, oxo-1,2-clihydropyridin-3- 3H), 2.79(s, 3H), 2.99(s, 3H) , 3.04(s, 3H) 323 yl)methyl)-1-(3- , 3.25-3.32 (m, 4H), 4.40 (brs, 111), 4.65-methoxybutan-2-y1)-2- 4.67 (d, J=5.6 Hz , 2H), 5.94(s, 1H). 7.49-methy1-6-(methy1sulfony1)- 7.51 (d, 1=8.4 Hz,1H), 7.61-7.63 (t, J=5.0 Hz, 1H-indole-3-carboxamide 1H), 7.98-8.00 (d, J=8.4 Hz, 1H), 8.119(brs, 1H), 12.71(brs, 1H) Compound Name NMR ink N-04-methoxy-6-methy1-2- (400 MHz, CDC13) 5 0.94 (d, J-7.2 Hz, 3H), oxo-1,2-dihydropyridin-3- 1.92(m, 3H), 2.19 (s, 3H), 2.75 (s, 3H), yOmethyl)-2-methyl-1-(3- 3.01(s, 3H), 3.90 (s, 4H), 4.65 (m, 2H), 4.87 460 (methylsulfonyl)butan-2- (m, 1H), 5.92 (s,1H), 7.10 (m, 2H), 7.38 (s, y1)-111-indole-3- 1H), 7.56 (d, J=5.2 Hz, 1H),7.89 (d, J=7.6 carboxamide Hz, 1H) (CDC13.
methoxy-6-methy1-2-oxo-400 MHz) 8 12.76 (s, 1H), 8.19-8.20 1-cyclopentyl-N-04-(d, J =2 .0 Hz, 1H), 8.11-8.13 (d, J =3 .6 Hz, 383 1 -dihydro idin-3-1H), 7.58 (s, 1H), 6.95-6.99 (m, 1H), 5.94 (s, yOmethyl)-2-methyl-lH-,2pyr 1H), 5.01-5.10 (m, 1H), 4.65-4.66 (d, J =5 .6 395 pyrro1 o[2 . - 3 hipyridine 3 - Hz, 2H), 3.90 (s, 3H), 2.82 (s, 3H), 2.39-2.42 (d, J=11.6 Hz, 2H),2.19 (s, 3H), 2.02-2.07(m, carhoxamide 3H), 1.72-1.75(m, 3H), 1.26-1.51(m, 1H) 1-cyclopentyl-N44,6-dimethy1-2-oxo-1,2-384 dihydropyridin-3-yOmethyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide ( )-N-((6-ethyl-4-methoxy- 'H NMR (400 MHz, CD3OD) 6 8.25-8.29 (m, 2-oxo-1,2-dihydropyridin- 2H). 6 7.28-7.31 (m, 1H). 6.89 (s, 1H), 4.93-/80 3-yl)methyl)-1-(1- 4.95 (br, 1H), 4.58 (s, 2H), 4.2-4.25 (m, methoxypropan-2-y1)-2- 1H),4.13 (s, 311), 3.77-3.81 (m, 1H), 3.24 (s, methyl-1H-pyrrolo[2,3- 311), 2.79-2.84 (q, 1H), 2.72 (s, 3H), 1.66-blpyridine-3-carboxamide 1.68 (d, J = 7.2 Hz, 3H) 1.32-1.36 (t, 311) (400 MHz, d6-DMS0) 8 11.57 - 11.65 (m, 1H), 8.18 - 8.23 (m, 1H), 8.07 - 8.12 (m, (R or S)-N-((4-Methoxy-6-1H), 7.83 - 7.91 (m, 1H), 7.07 - 7.15 (m, methy1-2-oxo-1,2-1H), 6.15 (s,1H), 4.31 (d, J = 4.46 Hz, 1H), dihydropyridin-3-288 4.04 - 4.20 (m, 1H), 3.88 - 3.97 (m, 1H), 439 y1)methy1)-2-methy1-1-(1-3.84 (s, 3H), 3.59 -3.70 (m, 1H), 2.97 -3.10 (tetrahydro-2H-pyran-4-(m 1H), 2.79 - 2.93 (m, 1H), 2.67 (br. S., yl)ethyl)-1H-pyrrolo[2,3- ' 3H), 2.20 (s, 3H), 1.78- 1.88 (m, 1H), 1.53 -b]pyridine-3-carboxamidc 1.68 (m, 3H), 1.28 - 1.41 (m, 2H), 0.97 -1.13 (m, 2H), 0.56 - 0.68 (in, 1H) h (400MHz, DMSO-d6) 8 11.82 - 11.68 (m, 1 (S)-N-((4-metoxy-6-H), 8.20 (dd, ./ - 1.6,4.7 Hz, 1 H), 8.13 (dd,./
methy1-2-oxo-1,2-= 1.6, 8.0 Hz, 1 H), 8.02 - 7.94 (m, 1 H), 7.33 dihydropyridin-3--7.26 (m, 2 H), 7.24 (d, J = 7.1 Hz, 1 H), 431 276 y pmethyl)-2-methyl-1 -(1-7.17 -7.10 (m, 3 H), 6.29 (s, 1 H), 6.19 (s, 1 rrolo[2phenylethyl)-1H-3-b]pyridine-3-H), 4.31 (br. S., 2 H), 3.83 (s, 3 H), 2.48 (hr.
py, S., 3 H), 2.22 - 2.18 (m, 3 H), 2.20 (s, 3 H), carboxamide 2.02 - 1.97 (m, 3 H), 2.00 (d, J = 7.4 Hz, 3 H) Compound Name NMR Ink (400MHz, DMSO-d6) 6 = 11.73 ¨ 11.56 (in, 1 H), 8.19 (d, J = 3.1 Hz, 1 H), 8.06 (dd, J =
(R or S)-N-((4,6-dimetbyl- 1.4, 7.9 Hz, 1 H), 7.82 (br. S., 1 H), 7.10 (dd, 2-oxo-1,2-dihydropyridin- J = 4.7, 7.8 Hz, 1 H), 5.91 (s, 1 H), 4.30 (br.
306 3-yl)methyl)-2-methyl-1- S., 2 H), 4.19 ¨ 4.02 (m, 1 H), 3.90 (d, J= 8.5 al., (1-(tetrahydro-2H-pyran-4- Hz, 1 H), 3.63 (d, J= 7.8 Hz, 1 H), 3.29 (s, 1 yl)ethyl)-1H-indole-3- H), 3.06 (s, 1 H), 2.92 ¨ 2.74 (m. 1 H), 2.64 carboxamide (br. S., 3 H), 2.25 (s, 3 H), 2.11 (s, 3 H), 1.80 (hr. S., 1 H), 1.59 (br. S., 3 H), 1.41 ¨ 1.24 (m, 1 H), 1.09 (s, 2 H), 0.67 ¨0.52 (m, 1 H) (CDC13, 400MHz) 6 8.14-8.12 (dd, J = 1.2 Hz,4.8 Hz, 1H), 8.09-8.07 (dd, J = 1.2 Hz,8.0 1-(4-amino-4-oxobutail-1-Hz, 1H), 7.51-7.46 (m, 1H), 6.94-6.91 (dd, .1 methyl-2-oxo-1,2-y1)-N-((4-methoxy-6-= 4.8 Hz,8.0 Hz, 1H),6.50 (s, 1H), 5.90 (s, 296 1H), 5.51 (s, 1H), 4.79-4.74 (dd, J = 6.4 411 dihydropyridin-3-Hz,14.8 Hz, 111), 4.44-4.39 (dd, = 4.8 yl)meth y1)-2-methyl-1H-Hz,14.8 Hz, 111), 3.87 (s, 311), 3.81-3.75 (m, pyrrolo[2,3-b]pyridine-3-1H), 2.83-2.78 (dd, J = J. Hz,14.8 Hz, carboxamide 1H),2.75 (s, 3H), 2.23 (s, 3H), 1.63-1.62 (d, J
= 6.8 Hz, 3H) (CDC13, 400MHz) 8 12.64 (s, 1H), 8.17-8.12 (-)-1-(1-c vanopropan-2-(m, 2H), 7.56 (s, 1H), 7.03-7.00 (dd, J = 4.4 y1)-N-04-methoxy-6-Hz,7.6 Hz, 1H), 5.94 (s, 1H), 4.89-4.80 (in, methy1-2-oxo-1,2-301 1H), 4.64-4.62 (d, J = 5.2 Hz, 1H), 3.90 (s, 3,4 dihydropyridin-3-3H), 3.74-3.68 (dd, J = 8.8 Hz,17.2 Hz, 1H), yi)methyl.)-2-methyl-lH-3.27-3.21 (dd, .1 = 6.4 Hz,16.8 Hz, 1H),2.79 pyrrolo[2,3-b]pyridine-3-(s, 3H), 2.19 (s, 3H), 1.78-1.76 (d, J = 6.8 Hz, carboxamide 3H).
(400 MHz, CDC13): 6 11.16 (br, 1 Fl), 8.19 (dd, 1 H, J1= 4.63 Hz, J2= 1.10 Hz), 8.06 (dd, ( )-N-06-(hydroxymethyl)-IF1, J1=7.49 Hz, J2 1.10 Hz), 7.14 (t, 1 H, 4-methoxy-l-oxo-12-dihydropyrid J=5.51 Hz), 7.05 (dd, 1 11, J1=7.49 Hz, 11, J2=

in-3-262 1)metby1)-1 4.63 Hz), 6.02 (s, 1H), 4.88-4.73 (br, 1 H), y-(1-4.57 (d, 2H, J=5.95 Hz), 4.47 (s, 2H), 4.22 (t, methoxypropan-2-y1)-2-1H, J= 8.60 Hz), 3.81 (dd, 1H, J1= 9.70 Hz, methy1-1H-pyrroloP,3-J2=3.8 Hz), 3.37-3.15 (br, 1f1), 3.21 (s, 3H), b]pyridine-3-carboxamide 2.76 (s, 3H), 2A4 (s, 3H), 1.64 (d, 3 H, J=7.06 Hz) Compound Name NMR Ink ( )-N-((4-methoxy-6-methy1-2-oxo-1,2-1 dihydropyridin-3-Racemic yOmethyl)-2-methyl-1-(1-268/269 (1-methy1-2-oxo-1,2-dihydropyridin-4-34)ethyl)-1H-pyrrolo[2,3-]pyridine-, 3-carboxamide (CDC13, 400MHz) 8 12.87 (brs, 1F1), 8.18-N-((4,6-dimethy1-2-oxo-8.11 (m, 2H), 7.56 (s, 1H), 7.47 (s, 1H), 7.01-1,2-dihydropyridin-3-7.00 (dd, J = 4.8 Hz, 8 Hz, 111), 5.96 (s, 1H), 2 71 yl)methyl)-1-(3-hydroxy-3-4.64-4.55 (in, 2H), 4.32-4.27 (dd, J 6.8 Hz 396 methylbutan-2-y1)-2-, 14 Hz, 1H), 2.73 (s, 3H), 2.42 (s, 3H), methy1-1H-pyrrolo[2,3-2.16 (s 3H), 1.52-1.50 (d, b]pyridine-3-carboxamide ' J =7.2 Hz,3H), (CDCI3, 400 M Hz) 8 11.92 (s, IF!), 8.21 (dd, ( )-N-04,6-dimethy1-2- J1 = 1.6 Hz, J2 = 4.8 Hz, 1H), 8.13 (dd, .11 =
oxo-1,2-dihydropyridin-3- 1.2 Hz, J. 2 = 7.6 Hz, 1H), 7.43 (t, J = 6.0 Hz, yOmethyl)-2-methyl-1-(1- 1H), 7.01 (dd, .11 = 4.8 Hz, .J2 = 8.0 Hz, 1H), 435 (piperidin-1-yl)propan-2- 5.94 (s, 1H), 4.60 (d, J= 6.0 Hz, 2H), 4.29 (d, y1)-1H-pyrrolo[2,3- J = 6.0 Hz, 2H), 3.13 (s, 1H), 2.80 (s, 3H), b]pyridine-3-carboxamide 2.66 (s, 2H), 2.43 (s, 5H), 2.20 (s, 3H), 1.44 (d, .1= 44.0 Hz, 611, 0.90 (dõI = 6.8 Hz, 311) ( )-N-(0,6-dimethy1-2- (400 MHz, Methanol-d4) 6:8.24-8.23 (m, oxo-1,2-dihydropyridin-3- 1H), 8.18-8.16 (m, 1H), 7.21-7.18 (m, I H), 193 yl)methyl)-1-(4- 6.37 (s, 111), 4.58 (s, 2 H), 3.51-3.45 (m, 1 H), 383 hydroxybutan-2-y1)-2- 3.49-3.15 (m, 2H), 2.74 (s, 4 H), 2.50 (s, 3 H), methy1-1H-pyrrolo[2,3- 2.34(s, 3 H), 2.31-2.26 (m, 1 H), 1.73-1.71 blpyridine-3-carboxamide (m, 3 H) N-((4,6-dimethy1-2-oxo-(400 MF1z, CDC13): 6 8.31-8.32 (d, 1H), 194 8.07-1,2-dihydropyridin-3-8.10 (d, 1F1), 7.38-7.40 (t, 1 11), 7.10-7.12 (t, yl)methy1)-2-methy1-1-1F1), 5.93 (s,111), 6.30 (s, 1F1), 4.56-4.57 (d, 458 (piperidin-1-y1su1fony1)- :
2H), 3.41-3.43 (t, 4H), 2.66 (s, 311), 2.40 (s, 1H-pyrro1o[2,3-b]pyridine-3-carboxarnide 3F1), 2.15 (s, 311), 1.47 (m, 611) ( )-N-((4,6-dimethyl-2-oxo- 1,2-dibydropyridin-3-209 yl)methyl)-1-(2-hydroxypropy1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxamide WO 2013/120104 PCT1tJS2013/025639 Compound Name 'H. NMR Ink ( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3- (400 MHz, Methanol-d4): 8 1.63 (d, J =6.8 254 yl)methyl)-4-(1- Hz, 3F1), 2.47 (s, 3H), 2.60 (s, 3H), 2.71 (s, methoxypropan-2-y1)-5- 3F1), 3.23 (s, 3H), 3.73-3.77 (m, 1171), 4.10-methyl-4H-pyrrolo[2,3- 4.15 (m, 1111), 4.60 (m, 2f1), 4.70-4.79 (m, dlthiazole-6-carboxamide 1.F1), 6.81 (s, 1.F1), 8.68 (s, 1.11) (400MHz ,DMSO-d6) 8 = 12.01 11.82 (in, 1 ( )-1-(3-methoxy-3- Fl), 7.91 7.82 (m, 2 HS), 7.71 - 7.64 (m, 1 methylbutan-2-y1)-N-04- H), 7.06 - 6.96 (m, 2 H), 6.25 (s, 1 H), 4.43 277 methoxy-6-methyl-2-oxo- (q, J = 7.1 Hz, 1 H), 4.33 (br. S., 2 H), 3.86 426 1,2-dihydropyridin-3- (s, 3 H), 3.14 - 3.09 (m, 3 H), 2.61 (s, 3 H), yl)methyl)-2-methyl-1H- 2.23 (s, 3 H), 1.58 - 1.52 (m, 3 H), 1.27 (s, 3 indole-3-carboxamide H), 0.88 (s, 3 H) (- )-N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-/75 yl)methyl)-1-(3-m.ethoxypentan-2-y1)-2-methyl-1 H-indole-3-carboxamide ( )-N-((4-methoxy-6-(CDC13, 400 M Hz) 8 7.85 (t, J = 6.4 Hz, methy1-2-oxo-1,2-1H), 7.45 (s, 211), 7.08-7.03 (m, 2F1), 5.93 (s, dihydropyridin-3-294 1E1), 4.71-4.61 (m., 211), 4.36 (s, 1F1), 3.90 (s, yl)methyl)-1-(3-4E1), 2.95 (s, 311), 2.75 (s, 3H), 2.17 (s, 3H), 412 methoxybutan-2-y1)-2-1.57 (d, J = 7.2 H7 3H) 1 /3 (d = 6 0 Hz J, = , =-= , = =
methy1-1H-indole-3-3E1) earboxamide ( )-N-((4-m.ethoxy-6-methy1-2-oxo-1,2- (400MHz, METHANOL-d4) S = 8.24 (d, dihydropyridin-3- J=8.0 Hz, 1H), 7.52 (d, J=8.5 Hz, 1H), 6.30 309 y1)methy1)-1-(1- (s, 1H), 4.94- 4.90 (m, 111), 4.54 (s, 2H), 4.36 467 methoxypropan-2-y1)-2- (t, J=9.5 Hz, 1H), 3.96 (s, 3H), 3.79 (dd, methyl-6-(trifluoromethyl)- J=4.8, 9.8 Hz, 1H), 3.22 (s, 3H), 2.73 (s, 3H), 1H-pyrrolo[2,3-b]pyridine- 2.34 (s, 3H), 1.70 (d, .1=7.0 Hz, 3H) 3-carboxamide N-04-methoxy-6-methy1-2-oxo-1, 2-dihydropyridin-3- (400 MHz, CDC13): 8 8.18-8.14 (m, 2H), yl) methyl)-2-methyl-1-(4, 7.57-7.51 (m, 1H), 7.05-7.02 (m, 1H), 5.92 (s, 4, 4-trifluoro-3- 1H), 5.01 (s, 2H), 4.63-4.60 (m, 1H), 3.89 (s, methoxybutan-2-y1)-1H- 3H), 3.73 (s, 3H), 2.73 (s, 3H), 2.20 (s, 3H), pyrrolo [2, 3-b] pyridine-3- 1.75-1.73(d, J=6.8, 3H.).
earboxamide Compound Name 'H NMR ink 1-(1-methoxy-2- (400 MHz, CDC13): 1.84 (s, 6H), 2.08 (s, rnethylpropan-2-y1)-N-(4- 2H), 2.86 (s, 3H), 3.24 (s, 3H), 3.82 (s, 2H), 338 methoxy-6-methy1-2-oxo- 3.88 (s, 3H), 4.65 (d, J=5.6 Hz, 2H), 5.88 (m, 1,2-dihydropyridin-3- 1H), 6.99-6.95 (m, 1H), 7.05-7.02 (m, 1H), yl)methyl)-2-methyl- III- 7.43-7.40 (m, 1H), 7.60-7.57 (m, 1H), 7.77-indole-3-carboxamide 7.75 (m, 1H), 12.98 (s, 1H) (400MFIz, DMSO-d6) 6 = 11.60 (br. s., 1 H), 7.72 (d, J = 7.6 Hz, 1 F1), 7.67 (d, .1 = 5.1 Hz, 2 H), 7.09 - 6.98 (m, 2 H), 6.14 (s, 1 HD, 4.41 ( )-1-(3-ethoxybutan-2-y1)-- 4.35 (m, 1 H), 4.32 (d, J = 4.9 Hz, 2 H), N-04-methoxy-6-methy1-2-290 4.03 - 3.93 (m, 1 H), 3.83 (s, 3 H), 3.25 (d, oxo-1,2-dihydropyridin-3- 426 = 9.4 Hz, 1 H), 2.82 - 2.72 (m, 1 H), 2.62 (br.
vl)methyl )-2-m ethyl-1H-' s 3 H), 2.19 (s, 3 H), 1.52 (d, J - 7.1 Hz, 3 indole-3-carboxamide H), 1.15 (d, = 6.0 Hz, 3 H), 0.68 (t, = 6.9 Hz, 3 H) (400 MHz, CDC13): 6 8.19-8.13 (m, 2H), N-((4-methoxy-6-methy1-2-7.57-7.55 (t, 1H), 6.99-6.96 (m, 1H), 5.94 (s, oxo-1, 2-dihydropyridin-3- -11-1), 4.67-4.65 (m, 2H), 4.40 (m, III), 4.16 293 yl) me methyl)-1-(3-(m, 111), 3.16 (s, 3H), 2.80 (s, 3H), 2.77 (s, 427 thoxypentan-2-v1)-2-' 3H), 2.20 (s, 311), 1.87-1.81 (m, 111), 1.67-methy1-1H-pyrrolo [2, 3-b]
1.65 (m,31I), 1.53-1.45 (m,311), 1.02-0.99 (m, pyridinc-3-carboxarnidc 3H) N-04-methoxy-6-methyl-2- (400 MHz, CDC13) 6 7.87-7.86 (d, 1H), 7.52- -oxo-1,2-dihydropyridin-3- 7.45 (m, 2H), 7.10-7.02 (m. 2H), 4.72-4.64 299 yl)methyl)-1-(3- (dd, 2H), 4.45-4.42 (s 1H), 3.9 (s, 3H), 3.73 4,5 methoxypentan-2-y1)-2- (s, 1H), 2.8-2.7 (d, 6H), 2.17 (s, 3H), 1.80-methy1-1H-indole-3- 1.75 (m,1H), 1.58 (s, 3H), 1.25 (m,1H), 1.03-carboxamide 0.99 (1, 3H) 1-(1-methoxy-1- (CDC13, 400MHz) 6 13.19 (s, 1H), 8.22-8.21 phenylpropan-2-y1)-N-((4- (d, J = 4.4 Hz, 1H), 8.16-8.14 (d, J = 8.0 Hz, 265 methoxy-6-methyl-2-oxo- 2H), 7.61-7.26 (m, 6H), 7.00-6.97(dd, .1 =
4.8 1,2-dihydropyridin-3- Hz,8.0 Hz, 1H), 5.94 (s, 1H), 5.33-5.30 (d, J

yl)methyl)-2-mcthyl-111- = 9.2 Hz, 1H), 4.73-4.63 (m, 2H), 4.48 (s, pyrrolo[2,3-b]pyridine-3- 1H), 3.90 (s, 311), 2.85 (s, 3H), 2.82 (s, 3H), carboxarnide 2.19 (s, 3H), 1.41-1.39 (d, J= 6.8 Hz, 3H) [003491 Exanwle 42. Synthesis of ( )-N4(4,6-diunethyl-2-oxo-1.2-thindrooµ
-I)methyl -2-rnethy 1-1 -0 43- e (Cotropound 205).
1003501 Step I: ( )-N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl-1-(1-(3-(4,4,5,5-tetramethy1-1,3,2-dioxaborolan-2-AphenyDethyl)-1H-indole-3-carboxamide:

es 4-ct ,04 )4.1 1 __________ t1P-Bs(Yt Z ====Cc\--1 tv 11,1 \lb ) ter I g I Airi \w/
A 50 mL round bottomed flask was charged with a magnetic stir bar, 1-(1 -(3-bromophenyl)ethyl)-N44,6-dimethyl-2-oxo-2,3-dihydropyridin-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide (171) (50 mg, 0.1 mmol), 4,4,4 ',4',5,5,5',5'-octamethy1-2,2'-bi(1,3,2-dioxaborolane) (38.7 mg, 0.15 mm.o1), potassium acetate (19.6 mg, 0.2 rrunol), 1, I%
Bis(diphenylphosphino)ferrocene palladium dichloride (110 mg, 0.15 mm.ol) and anhydrous 1,4-dioxane (5 mL) under N2. The reaction flask was fitted with a reflux condenser and the mixture was then heated to refluxed overnight. The reaction as allowed to cool to rt and the solvent was removed in vacuo and the resulting residue was purified by silica gel chromatography (Eluent:
PE/EA... 10:1) to the title compound as a yellow oil. (20 mg, yield 36 %) LCMS
(M + H-11 miz:
calc'd 491.12; found 492.3.
[003511 Step 2: ( )-N-04,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(3-(pyrimidin-4-y1)phenypethy1)-114-indole-3-carboxamide (Compound 205):
)1 )3 N
/2) a0 * .....
\/

A round bottom flask was charged with a magnetic stir bar, 141-(3-Bromo-pheny1)-ethyl]-2-methyl-IH-indole-3-carboxylic acid (4,6-dimethy1-2-oxo-2,3-dihydro-pyridin-3-ylmethyl)-amide (Step 8) (20 m.g, 0.04 mmol), 4-Chloro-pyrimidine (6.95 mg, 0.06 mmol), potassium carbonate (10.9 mg,0.08 mmol), 1,1%Bis(diphenylphosphino)ferrocene palladiumdichloride (44 mg, 0.06 mmol) and 1,4-dixoane1H20 (4:1, 8 mL). The mixture was purged and placed under under N2 and stirred heated to reflux with stirring overnight. The reaction was then allowed to cool to rt and the mixture was concentrated in vacuo. The resulting residue was purified by preparative-HPLC (Column: YMC ¨Actus Triart C18150*30mm *5um; Mobile phase A: water with 0.1 %
HCI solution; Mobile phase B: MeCN; column temperature: 30 C; Gradient: 35-65 % B). The collected fractions were combined and lyophilized to afford the title compound (6.3 mg, yield 35 %) LCMS (M + H+) calc'd 491.23; found 492.0; ill NMR (400 MHz, MeOD-d4 ö
9.2 (s, 1H), 8.85-8.54 (d, 2H), 8.14-8.06 (in, 2H), 7.78-7.72 (m, 1H), 7.58-7.54 (t, 1H), 7.44-7.42 (d,1H), 7.15-7.12(d, 1H), 7.04-7.01 (t, 2H), 7.01-6.79 (t, 1H), 6.8 (s,1H),6.14-6.12(t, 1H), 4.88(m, 2H), 4.67(s, 1H), 2.73(s, 3H), 2.6(s, 3H), 2.47(s, 3H), 2.08-2.07(d, 3H).
[003521 FxampIe 43. Synthesis of Methyl I -(1-(1A-d ox a n-2-yl)ethyll-2-methyl-1 H-indole-3-earboxylate. The title compound was used as an alternate alkyl carboxylate starting material in Step 3 of Example 36.
1003531 Step 1: 1-(1,4-dioxan-2-y1)ethanone:

Benzoyi peroxide To a solution of benzoic peroxide (20 g, 141 mmol) in 200 mL 1,4-dioxane at room temperature under nitrogen atmosphere was added biacetyl (24.3 g, 282 mmol). After the addition, the mixture was heated to reflux and stirred for 24 hours. The reaction mixture was cooled to 0 C.
The pH was adjusted to around 9 by progressively adding 2N sodium hydroxide below 0 C, extracted with 2-methoxy-2-methylpropane (10 mI, x 3), and concentrated to give 1-(1,4-dioxan-2-yl)etharione (13 g, 36%) as a yellow oil which was used directly in the next step without purification.
[003541 Step 2: 1-(1,4-dioxan-2-yl)ethanamine:
So F'' 1.) H2N

2.) H2, Pd/C 0 To a solution of 1-(1,4-dioxan-2-ypethanone (12 g, 92.2 mmol) in 1,2-dichloroethane (100 rni,) was added (4-methoxyphenypmethariamine (25 g, 184.4 mmol) at room temperature.
The mixture was allowed to stir for 3 hours, and then sodium triacetoxyborohydride (39 g, 184.4 mmol) was added. The resulting mixture was allowed to stir for 48 hours at room temperature.
The reaction mixture was quenched by adding water, extracted with dichloromethane (100 mL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel (elute:
dichloromethane/methanol 100:1-450: I -420:1) to give 1-(1,4-dioxan-2-y1)-N-(4-methoxybenzyl)ethanamine (16.4 g, 71%) as a yellow solid. LCMS (M Fl+) miz:
calcd. 251.15, found 251.9. To a solution of 1-(1,4-dioxan-2-yD-N-(4-methoxybenzyDethanamine (5 g, 19.9 mmol) in anhydrous methanol (100 m.L) was added palladium 10% on carbon (240 mg, 2 mmol), then purged with hydrogen (30 psi), the mixture was allowed to stir overnight at room temperature. The reaction mixture was filtered, and the filtrate was concentrated to afford the title compound (2.5 g, 96%) as a brown solid.
1003551 The amine intermediates shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure iniz I
tert-butyl 3-(1-aminoethyDpiperidin SocN NH2 228 e-l-carboxylate ( )-1-(4,4-r4H2 difluorocyclohexyDe 164 thanamine ---------(methylsulfonyl)azet 0, N "2 179 µs, idin-3-yDethanamine ( )-tert-butyl 4-(4- ri. NH2 (1- N
aminoethy1)154yridi N 307 ne-2-yOpiperazine-1-) carboxylate Bac 1003561 Step 3: (E)-methyl 34(1-(1,4-dioxan-2-371)ethyl)imino)-2-(2-bromophenyl)butanoate:
io 0 o Br (0)"1"N H2 _Dm. so Br To a solution of 1-(1,4-dioxan-2-yl)ethanamine (2.5 g, 19 mmol) in methanol (100 mL) was added methyl 2-(2-bromopheny1)-3-oxobutanoate (5.4 g, 20 mmol) and acetic acid (1.8 g, 30 mmol). The resulting reaction system was warm to reflux and allowed to stir overnight. The reaction mixture was concentrated and purified by column chromatographed on silica gel (eluted:

dichloromethane/methanol 50:1---+20:1--)5: I) the title compound (1 g, 14%) as a brown solid.
LCMS (M +11+) m/z: calcd. 383.07, found 384.9.
[003571 The imino-bromo intermediates shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials (e.g., one of the amines set forth in the table in Step 2 of this example) and modifications.
Name Structure mlz r, (E)-tert-butyl 3-(1-((3-(2- II`
bromophenyI)-4-methoxy-4- Boc N " (N C3-oxobutan-2- 482 si fir ylidene)amino)ethyl)piperidine-l-carboxylatc .....
"
p) '.1 r.L1( . ,...' (01-(4,4-difluorocyclohcxypethypitnino)butan F Br 417 oate Ui (E)-tert-butyl 4-03-(2-bromopheny1)-411:1 4-methoxy-4-oxobutan-2- B, ylidene)amino)piperidine-l-Boe carboxylate 0 -0-11%
_ O. Br (Z)-methyl 2-(2-bromophcny1)-3- N c) 398 (quinolin-5-ylamino)but-2-enoate IN; ...
(E)-methyl 2-(2-bromophenyI)-3- OS Br 339 (cyclopentylimino)butanoate 4r (E)-methyl 2-(2-bromophenyI)-3-((6-...
methylquinolin-5-yDimino)butanoate 10 412 N.

Br (- )-(E)-methyl 2-(2-bromopheny1)-3- ,=1%1... 11...
((1 -(1-(methylsulfonyl)azetidin-3- 432 ypethypimino)butanoate c, Name Structure rri/z ( )-(E)-tert-butyl 4-(4-((3-(2- Br bromopheny1)-4-inethoxy-4-oxobutan-2-ylidene)amino)pyridine-2-yDpiperazine-l-carboxylate Soc.
(E)-methyl 2-(2-bromopheny1)-3-((2,5-dimethy1pheny1)amino)but-2- 1-1 375 N
enoate (E)-methyl 2-(2-bromopheny1)-3-r ((2,3-dimethylphenyl)amino)but-2- 375 cnoatc 0 Cr (E)-methyl 2-(2-bromopheny1)-3-(quinolin-6-ylimino)butanoate 1003581 Step 4: Methyl 1-(1.-(1,4-dioxan-2-yOethy1)-2-methyl-1i-1-indole-3-carboxylate:

0),), Br RuPhos f Xs. 0 ====. 0 To a solution of (E)-methyl 3-((1-(1,4-dioxan-2-yflethyl)imino)-2-(2-bromophenyl)butanoate (400 mg, 1.1 mmol) in dioxane (3 mL) was added Chloro[2-(dicyclohexylphosphino)-3,6-dimethoxy-2',4',6'-triisopropylbiphenyl][2-(2-aminoethyl)phenyl]Pd( U) (160 mg, 0.2 mmol), 2-Dicyclohexyphosphino-2',6'-diisopropoxybiphenyl (93 mg, 0.2 mmol) and sodium tcrt-butoxide (192 mg, 2 mmol). The resulting reaction mixture was heated to 120 C with stirring for 30 mins in a microwave. The reaction mixture was quenched by adding water and was extracted with ethyl acetate (25 mL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel (eluted: petrol ether acetic ester 10:1-95:1-42:1) to afford the title compound (282 mg, 89%) as yellow solid. LCMS (M Fr) calcd. 303.15, found 303.9.

1003591 The compounds shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials (e.g., one of the imino-bromo intermediates shown in the table in Step 3 of this example) and modifications.
Name Structure miz =
\roq.loc .
methyl 1-(1-(1-(tert-butoxycarbonyppiperidin-3-ypethyl)- N
i 401 2-methyl-1H-indole-3-carboxylate = i 4 ( )-methyl 1-(1-(4,4- F
'4)(F
difluorocyclohexA N ethyl)-2-methyl- 336 ,,o 1 1H-indole-3-carboxylate . 4 o cioc:
methyl I -(1-(tert-butoxycarbonyl)piperidin-4-y1)-2- N 373 methy 1-1H-indo le-3-carboxy late I ...-c:, az 0 ii-IP v 1 Nµ
methyl 2-m ethy 1.-1-(ciu inoli n-5-y1)-1H-indole-3-carboxylate ,, I

methyl 1-cyclopenty1-2-methyl-1H- 9 indole-3-carboxylate 0 , I ,,, ...- µ

methyl 2-methyl- 146- = '', methylquinolin-5-y1)-1H-indole-3- N 331 I
carboxylate ..,1 o ( )-methyl 2-methyl-1-(1-(1- '--04-1-..., (methylsulfonypazetidin.-3-yflethyl.)- -, 351 At, 1H-indole-3-carboxylate ...=-=
liAP

cyc.
( )-methyl I -(1-(2-(4-(tert-butoxycarbonyl)piperazin-I -y1) pyridine-4-ypethyl)-2-methyl-1H- \)--indole-3-carboxylate o I i2ir .., Witfr c WO 2013/120104 PCT1tJS2013/025639 Name Structure ----------- rniz methyl 1-(2,5-dimethy1pheny1)-2-methyl-1H-indole-3-carboxylate methyl 1-(2,5-dimethylpheny1)-2-methyl-1H-indole-3-earboxylate methyl 2-methyl- I -(quinolin-6-y1)-I H-i ndole-3-carboxyl ate 41, 1003601 These alkyl carboxylates were also used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[003611 Example 44. Synthesis of ethyl 2-methy1-1-(1-(1-methy1-2-oxo-1.2-dihydropvtidin-4-v1)ethyl)-1H-indole-3-carboxylate.
\o Mel,A/V.*M000 IA\ *o 1114Ir lodomethane (57.94 mg, 0.408 mmol) was added to ( )-ethyl 1-(1-(2-methoxypyridin-4-Aethyl)-2-methy1-1H-indole-3-carboxylate (Example 39; 50 mg, 0.136 mmol). The mixture was stirred in the microwave at 150 C for 15 minutes. The mixture was evaporated to afford the title compound which was used without further purification (50 mg, yield:100%) as a starting alkyl carboxylatc in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[00362) Example 45. Synthesis of N-((4-nnthoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)meth1,1)-2-meth yl-1-(l-(1-(methylsolfonybRineridin-3-v1)ethyl)-111-in dole-3-ca rboxa mid e (Compound 318):

1 µ).....01 1 f-r:
\ e \
. õ
YX

''' 1 H 1 N
MsCI, Et3N . ..=== 1 1 i HN N alik hN 1 i, 1 4 0 0 11411Pr. 0 0 To a solution of N-((4-methoxy-6-methyl-2-oxo- I ,2-dihydropyridiii-3-yl)methyl)-2-methyl-1-( I -(piperidin-3-ypethyl)-111.-indole-3-carboxamide (20 mg, 45.81 pmol) in dichloromethane (3 mL) was added triethyl.amine (9.27 mg, 91.63 gmol) and methanesulfonyl chloride (7.87 m.g,68.72 Limo!). The mixture was stirred at room temperature for 12 hours. The. mixture was evaporated and purified by preparative-HPLC (Instrument: Gilson GX281 Column: Phenomenex Gemini C18 250*21.2mm Mobile phase A: water with 0.01molfINH4HCO3; Mobile phase B:
MeCN
Column temperature: 30 C Gradient: 23-53% B 10 min)to afford the title compound (7 mg, yield: 29.69%). LRMS (M + H+) miz: calc'd 515.22; found 515.2. 'H NMR (400 MHz, METHANOL-4) 8 0.94 - 1.12 (m, 2 H) 1.39 (br. S., 2 H) 1.57- 1.70 (m, 4 H) 2.33 (s, 3 H) 2.63 (s, 3 H) 2.83 (dõ/=9.54 Hz, 2 H) 2.88 (s, 3 H) 3.48 (br. S., 1 H) 3.79 - 3.87 (m, 1 11) 3.95 (s, 3 II) 4.38 (br. S., 1 Ft) 4.54 (s, 2 H) 6.28 (s, 1 H) 7.07 - 7.17 (m, 2 H) 7.58 (d, .1=8.03 Hz, 1 H) 7.72 (d, j=7.53 Flz, 1 H).
[003631 The compounds shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
The structure of these compounds is shown in Figure I.
Compound Name 'H NMR. , ink ill NMR (40011,111z, METHANOL-d4) 8 7.74 N-((4-methoxy-6-methyl- (d, .1=7.0 Hz, 1H), 7.65 - 7.54 (m, 1H), 7.17 -2-oxo-1,2-dihydropyridin- 7.08 (m, 2H), 6.30 (s, 111), 4.70 - 4.60 (m, 320 3-yl)methyl)-2-methyl-1- 1H), 4.56 (s, 2H), 4.34 - 4.24 (m, 1H), 3.97 0.-(1- (s, 3H), 3.80 (d, .1=13.6 Hz, 11), 3.19 - 3.01 479 (methylsulfonyl)piperidin- (m, 1H), 2.77 - 2.69 (m, 1H), 2.64 (s, 3H), 3-yl)ethyl)-1H-indole-3- 2.56 - 2.41 (m, 1H), 2.35 (s, 3H), 2.26 -2.12 carboxamide (m, 31), 1.73 - 1.63 (m, 3H), 1.62 - 1.50 (m, 1H), 1.35- 1.23 (m, 1H), 1.09 (s, 2H) N-((4-methoxy-6-methyl- (CDC13, 400 M Hz) 5 7.94 (d, J = 7.6 Hz, 2-oxo-1,2-dihydropyridin- 1H), 7.88 (d, J= 8.0 FIz, I H), 7.63 (d, .1 = 6.0 371 3-yOmethyl)-2-methyl-1- Hz, 1H), 7.36 (t, J = 8.0 Hz, 1H), 7.16-7.01 (1-(methylsulfony1)- (m, 3H), 6.78 (d, J = 7.6 Hz, 1H), 5.97 (s, 1,2,3,4-tetrahydroquinolin- 1H), 4.70 (t, J = 19.6 Hz, 2H), 3.92 (s, 3H), 5-y1)-1H-indok-3- 3.78 (d, J = 5.6 Hz, 2H), 3.00 (s, 31), 2.49 (s, carboxami de 3H), 2.32 (s, 3H), 2.18-2.13 (m, 2H), 1.85-1.81 (m, 2H) [00364] Example 46. Chiral Separation of Compound 219 to afford Compounds 223 and 224. N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-ypmethyl)-1-(1-methoxypropan-2-y1)-2-methyl-1H-indole-3-carboxamide (200 mg) (Compound 219) was subjected to chiral chromatography via supercritical fluid chromatography (SFC) (A:C2H5OH,B:NH3=H20.
A:B=55:45 AD column) to afford the separate enantiomers 223 (peak 1) and 224 (Peak 2) (60 mg each) LCMS 398 (M-Fl)' 1H NMR (400 MHz, CD30D) 6 7.69 (d, J=7.2 Hz,1H), 7.53 (d, J=7.6 Hz,1H), 7.12 (m, 2H), 6.26(s,1H), 4.80(m,1H), 4.52(8,2H), 3.99 (m, 4H),3.75 (m, 1H),3.20 (s, 3H), 2.62 (s, 3H), 2.31 (s, 3H), 1.59 (d, J=7.2 Hz, 3H). The optical rotation of each enantiomer was not determined.
[00365] The compounds shown in the following table were prepared according to the general chiral chromatography procedure outlined above. The optical rotation of the separated enantiomers was not determined, but the elution peak ("Peak 1" or "Peak 2") is indicated.
Structures of each compound are shown in Figure 1.
Compound Name H NMR tniz (R or S)-N-((4,6-dimethy1-2- (4)0 MHz, CD30D) 6 7.74 (m, III), oxo-1,2-dihydropyridin-3- 7.57 (d, J=7.6 Flz,1H), 7.15 (m, 2H), 6.14 217 yl)methyl)-1-(1- (s,111), 4.86 (m,11-1), 4.55 (8,211), 4.02 methoxypropan-2-y1)-2- (m, 1H), 3.77 (m, 1F1), 3.22 (s, 3H), 2.65 methyl-1H-indole-3- (s, 3H), 2.43 (s, 3H), 2.26 (s, 3H), 1.62 carboxamide-PEAK 1 (d, 3=7.2 Hz, 3H) (R or S)-N-((4,6-dimethy1-2- (400 MHz, CD30D) 6 7.74 (m, 1H), oxo-1,2-dihydropyridin-3- 7.57 (d, J=7.6 Hz,1H), 7.15 (m, 2H), 218 yl)methyl)-1-(1- 6.14(s,1H), 4.86(m,1H), 4.55(s,2H), 4.02 methoxypropan-2-y1)-2- (m, 1H),3.77 (m, 1H),3.22 (s, 3H), 2.65 methy1-1H-indole-3- (s, 3H), 2.43 (s, 3H), 2.26 (s, 3H), 1.62 carboxamide-PEAK 2 (d, J=7.2 Hz, 3H) (400 MHz, Methanol-d4) 67.84 (d, (R or S)-1-(1-(3-carbamoylphenyflethyl)-N-J=8Hz,1H), 7.66 (d, j=8.4 Hz, 2H), 7.33-7.37 (m, 2H), 7.04 (m, 1 H), 6.88-6.96 225 ((4,6-dimethy1-2-oxo-1,2-(m, 2H), 5.90 (s, 2H),5.81-5.82 (m, 1 H), 457 dihydropyridin-3-yOmethyl)-2-4.57-4.63 (m, 2 H), 3.54 (s, 1 H), 2.99(s, methy1-1H-indole-3-3H), 2.70(s, 3H), 2.45(s, 3H), 2.22(s, carboxamide PEAK 1 3H), 1.96(d, J=7.2 Hz, 3H) Compound Name 'H NMR
(400 MHz, Methanol-d4) V.84 (d, J=8Hz,1H), 7.66 (d, J=8.4 Hz, 2H), 7.33-carbamoylphenypethyl)-N-7.37 (m, 2H), 7.04 (m, 1 H), 6.88-6.96 226 04,6-dimethy1-2-oxo-1 (m, 2H), 5.90 (s, 2H),5.81-5.82 (m, 1 H), 457 dihydropyridin-3-yl)methyl)-2-4.57-4.63 (m, 2 H), 3.54 (s, I H), 2.99(s, methy1-1H-indole-3-3H), 2.70(s, 3H), 2.45(s, 3H), 2.22(s, carboxamide PEAK 2 3H), 1.96(d, J=7.2 Hz, 3H) (R or S)-1-(1-(3- (400 MHz, Methanol-d4) 6 7.78 (d, eyanophenypethyl)-N-04,6- J-8Hz, 1H), 7.67 (d, J=7.2Hz,1H), 7.54 227 dimethy1-2-oxo-1,2- (m, 3H), 7.13 (m, 1H), 7.03 (d. J::4 Hz, 2 dihydropyridin-3-yl)methyl)-2- H), 6.45 (s, 1H), 6.04 (m, 1F1), 4.61 (s, methyl-111-indole-3- 2H), 2.66 (s, 3 H), 2.52 (s, 3 IT), 2.36 (s, carboxamide PEAK 1 1 H), 2.02 (d, J=7.2 Hz, 3H) (R or S)-1-(1-(3- (400 MHz, Methanol-d4) 6 7.78 (d, cyanophenyflethyl)-N-((4,6- J=8Hz, 1H), 7.67 (d, J=7.2Hz,IH), 7.54 228 dimethy1-2-oxo-1,2- (m, 3H), 7.13 (m, 1H), 7.03 (d, J=4 Hz, 2 dihydropyridin-3-yl)methyl)-2- H), 6.45 (s, 1H), 6.04 (m, 1H), 4.61 (s, methy1-1H-indole-3- 2H), 2.66 (s, 3 H), 2.52 (s, 3 H), 2.36 (s, carboxamide PEAK 2 1 H), 2.02 (d, J=7.2 Hz, 3H) (400 MHz, CD30D) 6 8.10 (brs, 1H), 8.06-8.04 (d, .1=8.0 Hz, 1H), 7.26-7.24 (R or S)-(4,6-dimethy1-2-oxo-(d, ./=8.0 Hz, 1H), 7.18-7.14 (t, J=7.6 Hz, 1,2-dihydropyridin-3-213 1H), 7.08-7.02 (m, 2H), 6.69-6.67(d, yl)methyl 1-(2,3-dihydro-1H- J=6.8 Hz, 1H), 6.00(s, 1H), 4.41(s, 2H), 427 inden- I -y1)-2-methy1-1H-2.74-2.64(m 2H), 2.46(s 3H) 2.30(s indole-3-carboxylate PEAK 1 ' " ' 3H), 2.17-2.11 (m, 1H), 1.52-1.49(m, 2H) (400 MHz, CDC13) (R or S)-1-(sec-buty1)-N-04,6-8 8.116-8.132 (d, 111), 8.011-8.035 (d, dimethy1-2-oxo-1,2-1H), 7.037-7.057(t, 1H), 6.056 (s, 1H), 195 dihydropyridin-3-yl)methyl)-2-4.462 (d, 2H), 6.30 (s, 1H), 2.63(s, 3H), 368 methy1-1H-pyrrolo[2,3- 2.356 (s 3H) 2.189(s 3H) 1.880-1.933 blpyridine-3-carboxamide " "
(m, 1H), 1.587-1.605 (d, 2H), 1.226 (s, 2H), 0.658 (t, 3H) (400 MHz, CDC13) (R or S)-1-(sec-butyl)-N-((4,6-6 8.116-8.132 (d, 1H), 8.011-8.035 (d, dimethy1-2-0x0-1,2-IH), 7.037-7.057(t, 1H), 6.056 (s, I H), 196 dihydropyridin-3-yOmethyl)-2-4.462 (d, 2H), 6.30 (s, 1171), 2.63(s, 3H), 368 methy1-1H-pyrrolo[2,3-2.356 (s 3H) 2.189(s, 3H), 1.880-1.933 b]pyridine-3-carboxamide (m, 1H), 1.587-1.605 (d, 2H), 1.226 (s, 21-1), 0.658 (t, 3H) Compound Name 'H NMR
(R or S)- ( )-141- NMR (400 MHz, CDC13):
cyclopropylethy1)-N4(4,6- 8 8.32-8.34 (d, 111), 8.18-8.2 (d, 1H), dimethy1-2-oxo-1,2- 7.27-7.30(m, 1H), 6.70 (s, 1H), 4.47 (s, dihydropyridin-3-Amethyl)-2- 2H), 3.94-3.95 (d, 1H), 2.61 (s, 3H), 2.43 379 methy1-1H-pyrrolo[2,3- (s, 3H), 2.29-2.30 (s, 3H), 1.57-1.59 (d, bipyridine-3-earboxamide 3H), 0.63-0.64 (t, 1H), 0.27-0.64 (m, PEAK 1 2H), 0.02-0.04 (t, 1H) (R or S)- ( )-141- NMR (400 MHz, CDC13):
eyelopropylethyl)-N4(4,6- 8 8.32-8.34 (d, 1H), 8.18-8.2 (d, 1H), dimethy1-2-oxo-1,2- 7.27-7.30(m, 1H), 6.70 (s, 114), 4.47 (s, dihydropyridin-3-yl)methyl)-2- 2H), 3.94-3.95 (d, 1H), 2.61 (s, 3H), 2.43 379 methy1-1H-pyrrolo[2,3- (s, 311), 2.29-2.30 (s, 3H), 1.57-1.59 (d, bipyridine-3-carboxamide 3H), 0.63-0.64 (t, 1H), 0.27-0.64 (m, PEAK 2 2H), 0.02-0.04 (t, 1H) (400 MHz, CDC13) 8 13.23 (s, 111), (R or S)-N-((4-methoxy-6-8.16-8.17 (m, 111), 8.11-8.13 (m, 111), methy1-2-oxo-1,2-7.57-7.60 (t, J = 5.211z, 1H), 6.93-6.96 dihydropyridin-3-yl)methyl)-1-256 (m, 1H), 5.92 (s, 1H), 4.82-4.83 (d, = 39, (1-methoxypropan-2-y1)-2-2.4 Hz, 11T), 4.65-4.66 (d, J= 6.4 Hz, methy1-1H-pyrrolo[2,3-2H), 3.89 (s, 311), 3.81-3.85 (m, 111), blpyridine-3-carboxamide 3.22 (s, 311), 2.79 (s, 3H), 2.17 (s, 3H), 1.64-1.66 (dõ/ = 8.0 Hz, 311) (400 MHz, CDC13) 8 13.23 (s, 1H), (R or S)-N4(4-methoxy-6-8.16-8.17 (m, 1H), 8.11-8.13 (m, 1H), methy1-2-oxo-1,2-7.57-7.60 (t, J = 5.2Hz, 1H), 6.93-6.96 dihydropyridin-3-yl)methyl)-1-257 (m. 114) 5.92 (s, 1H), 4.82-4.83 (d, J = 399 (1-methoxypropan-2-y1)-2- "
2.4 Hz, 1H), 4.65-4.66 (d, J = 6.4 Hz, methy1-1H-pyrrolo[2,3-2H), 3.89 (s, 3H), 3.81-3.85 (m., 111), bipyridine-3-carboxamide 3.22 (s, 3H), 2.79 (s, 3H), 2.17 (s, 311), 1.64-1.66 (d, 8.0 Hz, 311) (400 MHz, METHANOL-d4) 8 ppm 1.77 (R or S)- N4(4-methoxy-6-(d, J=6.84 Hz, 3 H) 2.34 (s, 3 H) 2.39 (s, methy1-2-oxo-1,2-3 H) 2.75 (s, 3 H) 3.64 (dd, J=14.66, 3.64 dihydropyridin-3-yl)methyl)-2-278 Hz, 1 H) 3.96 (s, 3 H) 4.54 (s, 2 H) 5.21 methyl-141-(br. S., 2 H) 6.30 (s, 1 H) 7.16 (dd, (rnethylsulfonyl)propan-7-v1)-J=7.94, 4.85 Hz, 1 H) 8.10 (dd, J=7.94, 1H-indole-3-earboxamide 1.54 Hz, 1 H) 8.27 (dd, J=4.63, 1.54 Hz, 111) Compound Name 'H NMR
(R or S)- N-((4-methoxy-6-(400 MHz, METHANOL-4) 8 ppm 1.76 methy1-2-oxo-1,2-(d, J=7.06 Hz, 3 H) 2.33 (s, 3 H) 2.37 (s, dihydropyridin-3-yl)methyl)-2- 3 H) 2.74 (s, 3 H) 3.62 (dd, J=14.66, 3.64 279 methy1-1-(1-Hz, 1 H) 3.94 (s, 3 H) 4.52 (s, 2 H) 5.19 (methylsulfonyl)propan-2-y1)- = '' 446 (br. S 2 H) 6.28 (s, 1 H) 7.15 (dd, 111-indole-3-carboxamide J=7.94, 4.85 Hz, 1 H) 8.09 (dd, J=7.94, PEAK 2 0.77 Hz, 1 H) 8.25 (dd, J=4.85, 1.54 Hz, 1 H) (R or S)-N4(4-methoxy-6-methy1-2-oxo-1,2- (400 MHz, CDC13) 8 ppm 1.90 (d, dihydropyridin-3-yl)methyl)-2-J=7.28 Hz, 3 H) 2.26 (s, 3 H) 2.61 (s, 3 268 methyl-1-(1-(1-methyl-2-oxo- H) 3.49 (s, 3 H) 3.91 (s, 3 H) 4.65 (d, 1,2-dihydropyridin-4-yl)ethyl)-1=5.51 Hz, 2 H) 5.72 (dd, J=7.06, 1.76 462 1H-pyrrolo[2,3-b]pyridine-3- Hz, 1 H) 5.96 (s, 1 H) 6.47 (s, 1 H) 7.06 carboxamide - 7.14 (m, 2 H) 7.61 (br. S., 1 H) 8.18 -PEAK I 8.28 (m, 2 H) (R or S)-N-((4-methoxy-6-methyl-2-oxo-1,2---(400 MHz, CDC13) 8 ppm 1.90 (d, dihydropyridin-3-yl)methyl)-2- ' .1-7.28 Hz 3 H) 2.26 (s, 3 H) 2.61 (s, 3 269 methyl-1-(1-(1-methy1-2-oxo- H) 3.49 (s, 3 H) 3.91 (s, 3 H) 4.65 (d, 1,2-dihyd.ropyridin-4-ypethyl)-J=5.51 Hz, 2 H) 5.72 (dd, J=7.06, 1.76 462 1H-pyrrolo[2,3-b]pyridine-3- Hz, 1 H) 5.96 (s, 1 H) 6.47 (s, 1 H) 7.06 carboxamide - 7.14 (m, 2 H) 7.61 (br. S., 1 H) 8.18 -PEAK 2 8.28 (m, 2 H) Trans-(R or S, R or S)-N-((4-methoxy-6-methy1-2-oxo-1,2-(CDC11 400 M Hz) 5 7.85 (t, J - 6.4 Hz, dihydropyridin-3-Amethyl)-1- III), 7.45 (s, 214), 7.08-7.03 (m, 2H), 307 (3-methoxybutan-2-y1)-2-5.93 (s, 114), 4.71-4.61 (m, 2I-1.), 4.36 (s, 411 methyl-1H-indole-3- Ili), 3.90 (s, 411), 2.95 (s, 3H), 2.75 (s, carboxamide 3H), 2.17 (s, 3H), 1.57 (d, J = 7.2 Hz, PEAK 1 3H), 1.23 (d, .1= 6.0 Hz, 3H) Trans-(R or S. R or S)-N4(4-methoxy-6-methy1-2-oxo-1,2- (CDCb, 400 M Hz) 5 7.85 (t,J = 6.4 Hz, 308 dihydropyridin-3-yl)methyl)-1-1H)' 7.45 (s, 2H), 7.08-7.03 (m, 21), (3-methoxybutan-2-y1)-2-5.93 (s, 1H), 4.71-4.61 (in, 2H), 4.36 (s, methyl-1H-indole-3- 1H), 3.90 (s, 4H), 2.95 (s, 3H), 2.75 (s, carboxamide 3H), 2.17 (s, 3H), 1.57 (d, J = 7.2 Hz, PEAK 2 3H), 1.23 (d, J = 6.0 Hz, 3H) N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methy1-141-phenylethyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxamide Compound Name 1H NMR , ink .
N-((4,6-dimethy1-2-oxo-1,2-. dihydropyridin-3-yOmethyl)-2-methy1-1-(1-phenylethyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxamide 1003661 Example 47. Synthesis of I -(1 -(3-cyanoonen vbethvb-N-((4.6-dimei hi, 1-2-0 KO- 1,2-dihydropyridin-3-0)methyll-2-ineths1-111-1ndo1e-3-earboxamide (Compound 187).
Br CN
--- \ / Zn(CN)2, Pr.VPhOzi Ni-----6 fit I N

1 -(1-(3-bromoplieny pet hyl.)-N44,6-dimethyl-2-ox o-1,2-dihy dropyridi n-3-yl)methyl)-2-methyl-1H-indole-3-carboxamide (Compound 171; 100mg, 0.2 mmol), Zn(CN)2 (36mg, 0.3 mmol), Pd(PPh3)4 (47 mg, 0.04 mmol) were combined in 2 ml DMF, then stirred at 100 C
for 30 min under N2 in mw. After the complete of the reaction, purified by pre-HPLC (A:
CH3CN, B:water-1-0.1%Ha. A:B=32:62 ASH CI8 150*25mm) to afford the title compound (35 mg, yield: 40%).1H NMR (400 MHz, Methanol-d4) V.78 (d, J=8 Hz, 1.H.), 7.67 (d, J=7.2Hz, Ill), 7.54 (m, 3H), 7.13 (m, 1H), 7.03 (d, .11=4 Hz, 2 H), 6.45 (s, 1H), 6.04 (m, 11-1), 4.61 (s, 2H), 2.66 (s, 3 H), 2.52 (s, 3 H), 2.36 (s, 1 H), 2.02 (d, j=7.2 Hz, 3H).
[00367] Example 48. Synthesis of ( )-1-(sec-butv1)-N-(01-methoxv-6-Inettiv1-2-oxo-1,2-dihydrouvridin-3-0methyl)-2-tvaet h% 14341) v ri dine-3-1,1)-1H-indole-3-carbox amide, (Compound 388).
\rj _N
i \,J(HO)A-0 A solution of ( )-1-(sec-buty1)-6-chloro-N-((4-methoxy-6-inethyl-2-oxo-1,2-dihydropyridin-3-yOmethyl)-2-methyl-IH-indole-3-carboxamide (Compound 185) (50mg, 0.12mmol), pyridine-3-ylboronic acid (22 mg, 0.18 mmol), tricyclohexylmethane (7mg, 0.024urno1), K3PO4 (120mg, 0.60 mmol), Tris(dibenzy1ideneacctone)dipa11adium(0)(10 mg), water (1 mL)and 1,4-dioxane (4 inL) was stirred under N2 at 100 C for 16 h. The reaction was then allowed to cool to rt and the mixture was diluted with water and dichloromethane, the aqueous layer was extracted with dichloromethane. The organic layers were dried with Na2SO4 and evaporated. The residue was purified by preparative-HFLC (Instrument: Gilson 215; Column: Gemini C18 10u I50*25mm;
Mobile phase A: Water(0.0225% ]Cl v/v); Mobile phase B:
Acetonitrile(neutral);Gradient:42-62(13%);Flowrate:25mUmin) The collected fractions were combined and lyophilized to give 1-(sec-buty1)-N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yOmethyl)-2-methyl-6-(pyridine-3-y1)-1H-indole-3-carboxamide as a yellow solid (3.2 mg, yield 5.7 %).LCMS (M
H+) rniz: calcd 458.23; found 459Ø 1H NMR (400 MHz, CD30D): 89.25 (s, 1H), 8.98 (d, J=8.4, 1H), 8.80 (d, J=5.6, 1H), 8.18 (t, J=5.6, 1H), 8.05 (s, 1H), 7.97(d, J=8.0, 1H),7.60(d, J=8.4, 1H), 6.86(s,1H), 4.64(m,1H), 4.62 (s,2H), 4.13 (s, 3H), 2.73(s, 3H), 2.53(s, 3H), 2.30(m, IH), 2.03-2.10(m, 1H), 1.71(d, J=6.8, 3H), 0.77(t, J=7.6, 3H).
[003681 The compounds shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
Structures are shown in Figure 1.
Compound Name 'FI NMR miz (400 MHz, CDC13) 8 7.77 (d, J=8.4, 1.11), (A-.)-1-(sec-buty1)-N-( (4-7 68 (s 11I) 7.60 (d 1=7 6 2H) 7.43-7.48 methoxy-6-methyl-2-oxo- '' ' ' (m, 3FI), 7.35 (t, J=7.6, 1H), 7.20(s, 389 1,2-dihydropyridin-3-1H) 6.31(s,11-1), 4.6/ (s,211), 4.49 (s, III), 458 y1)methyl)-2-methy1-6-3 95' (s 311), 2.76 (s. 311), 2.67 (s, 31-1), pheny1-11I-indole-3-2.18-2.24 (m, III), 1.94-2.01 (m, I EI), 1.64 earboxamidc J=7.2, 3141 . 0.76 0, J=7.6, 31-I) (400 MHz, CDC13) 8 0.74-0.780, J=7.2 Hz, 1-(sec-buty1)-N-44-3H), 1.59-1.61(d, J=6.8 Hz, 3H) ,1.88-( )-1.98(m, 1H),2.18 (s, 5H), 2.74 (s, 3H), 3.02-methoxy-6-methy.1-.2-oxo-3.04(t, J=5.0Hz, 3H), 3.55-3.58(t, J=5.2Hz, 1,2-dthydropyndm-3-349 3H) 3.85-3.86(d, J=4.4 Hz, 3H),4.45 (brs, yl)methyl)-2-methy1-6-(6-1H), 4.66-4.68(d, J=5.6Hz, 2H),5.89 (s, (piperazin- I -yl)pyridine-3-1H), 6.62-6.65(d, J=8.8Hz, 1H),7.15-7.17 y1)-1H-indo le-3-(t. J=7.6Hz, 1H), 7.54(s, 1H),7.64-7.67 (m, carboxamide 2H), 7.85-7.87(d, J=8Hz, I H), 8.42-8.43(d, J=2.8Hz, 11-1) 1003691 Example 49. Synthesis of 2-methyl- I -11,2,3,4-tetrahvdrouuinolin-5-vI)-1 indole-3-earboxvlie acid.

-.N NH
\ =
H2. Pt02 HO alla HO II

To a solution of 2-methyl-1-(quinolin-5-y1)-1H-indole-3-carboxylic acid (Example 40) (25 mg) in acetic acid (3 mi.) was added Pt02 (50 mg) at 22 C. The reaction was stirred at 22 C under H2 ball for 20 hrs. Then the mixture was filtered and the filtrate was purified by column chromatography on silica gel eluted with dichloromethane: methanol = 70:1 to the title compound as a yellow solid (20 mg, 71%).
[00370] The compounds shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting alkyl carboxy late.
Name Structure miz 2-methyl-1-(6-methy1-1,2,3,4- H
tetrahydroquinolin-5-y1)-1H-indole- 311 3-carboxylic acid HO i 0 liar ccH
2-methyl-1-(1,2,3,4-tetrahydroqui nolin-6-y1)-1H-indole- 307 3-carboxylic acid Ho r.
1003711 The carboxylic acids of this example were used as starting material in Step 4 of Example 36 in the synthesis of certain compounds of the invention.
[003721 Example 50. Synthesis of 2-metbv1-1-(3-methvlbutano% 1)- 1 ndole-3-carbox vile acid. The title compound was used as starting material in Step 4 of Example 36 in the Sy11111C:-.1:-.
of certain compounds of the invention.
[003731 Step 1: tert-butyl2-methyl-1-(3-methylbutanoy1)-1H-indole-3-earboxylate = NH
14) CI
To a solution of tert-butyl 2-methyl-1H-indole-3-carboxylate (300 mg, 1.30 mmol) in N,N-dimethylformamide (10 mL) was added sodium hydride (103.76 mg, 2.59 mmol, 60%). The mixture was stirred at 0 C for 30 minutes. Then 3-methylbutanoyl chloride (234.60 mg, 1.95 mmol) was added at 0 C and the mixture was stirred at room temperature(25 C) for 2 hours. The mixture was cooled with ice, quenched with water (5 mL), diluted with ethyl acetate (20 mL), washed with brine (20 mL), extracted with ethyl acetate three times (20 mLx3), then dried over sodium sulfate. After concentration, the residue was purified by column on silica gel (0%-5%
PE/EA) to afford the title compound (200 mg, yield: 48.89%).
[003741 The compound shown below was prepared according to the general procedure outlined in above using the appropriate starting materials and modifications.
Name Structure raiz o tert-butyl 2-methy1-1-(tetrahydro-2H-t.4 pyran-4-carbony1)-1H-indole-3- = / 1 344 carboxylate 1 411 0037.51 Step 2: 2-methyl-1-(3-methylbutanoy1)-1H-indole-3-carboxylic acid:
0,Y
TM ¨N
>r0 f g To a solution of tert-butyl 2-methyl-1-(3-methylbutanoy1)-1H-indole-3-carboxylate (200 mg, 634.10 mol) in dichloromethane (5 mL) was added trifiuoroacetic acid (5 mL).
The mixture was stirred at room temperature (25 C) for 1 hour. The mixture was evaporated and partitioned between dichloromethane and water. The aqueous layer was washed with dichloromethane (10 mLx2), combined the organic layers, dried over sodium sulfate, then filtered and concentrated in vacuum afford the title compound (30 mg, yield:89%).
[003761 The carboxylic acids shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure m/z 2-methy1-1-(tetrahydro-2H-pyran-4-carbonyl)-1H-indole-3-carboxylic Ho / 288 acid ( )-1-(2,3-dihydro- I H-inden-l-y1)-2- 111.
methy1-1H-pyrrolo[2,3-h]pyridine-3- H 292 carboxylic acid 4 o Name Structure rri/z ..o ( )-2-methyl-1-(tetrahydrofuran-3- ' y1)-1H-pyrrolo[2,3-13]pyridine-3- );===N.

carboxylic acid 0¨

( )-1-(1-mcthoxypropan-2-y1)-2-methy1-1H-pyrrolo[2,3-13]pyridine-3- 249 carboxylic acid HgL
\
\c) ( )-1-(1-(3-methoxyphenyl)ethyl)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3- 311 carboxylic acid Flo,t1).1 \o ( )-1-(1-(2-meth oxypyridi n-4- rr-4 yl)ethyl)-2-methy1-1H-pyrrolo[2,3- N 312 b]pyridine-3-carboxylic acid 1-(chroman-4-y 1)-2-methy1-1H-;.) pyrrolo [2,3-b]pyridine- 3-carboxylic r *Ni `2--=/ 309 acid N
( )-1-(1-cyclopropylethyl)-2-methyl-1. H-pyrrolo[2,3-b]pyridine-3-¨N 245 carboxylic acid ( )-1-(1-ethoxypropan-2-yI)-2-methy1-1H-pyrrolo[2,3-11pyridine-3- NrN 263 carboxylic acid HO
-11"Lsi ( )-1 -(1-cyanoethyl)-2- methyl-1H-pyrro lo [2,3-b]pyridine-3-carboxylic HO 230 acid \r) oxypyridi n-3-yl)ethyl)-2-methyl- I H-pyrrolo[2,3- N 312 b]pyridine-3-carboxylic acid Ho Name Structure rri/z ( )-2-methy1-1-( I -phenylpropy1)-111- *
pyrrolo [2,3-b]pyri din e-3-carboxy I i c NJ 295 acid HO 1 \ --1/4 <7 1 1 N-N
( )-1-(1-(1H-pyrazol-1-yl)propan-2-µ)--i y1)-2-methyl-1H-pyrrolo[2,3- syctdi . NI 285 b]pyri di ne-3-carboxy I ic acid o "b r47.-.4 ( )-1-(1-(2-methoxypyrimidin-4-ypethyl)-2-methyl-1H-pyrrolo[2,3- N 313 b]pyridine-3-carboxylic acid µ 1 ( )-2-Methyl.- 14 T
1- µ ,\ 4-) morpholinopropan-2-y1)-1H- )---; 304 pyrrolo[2,3-b)pyridine-3-carboxylic N
acid µ
0 , ( -)-1-(1-(IH-benzo[d] imidazol-1-yl)propan-2-y1)-2-methyl- I H-pyrrolo[2,3-b]pyridine-3-carboxylie 110,1õ..k.b acid ci i= \
N
Ii ( )- # 1-(1-(3-cyanophenyl)ethyl)-2-methyl-1H-pyrrolo[2,3-b]pyridinc-3- '..,,,; 306 carboxylic acid hoyll-t--) ________________________________________ 0 \f, I -(3-methoxybutan-2-yI)-2-methyl-1H-pynolo[2,3-bipyridine-3- ===..,..N 263 HO,TrolLb carboxylic acid \ /

CON H, H-1-(1-(3-carbamoylphenypethyl)-ft 2-methyl-I H-pyrrolo[2,3-b]pyri dine- 324 3-carboxylic acid Ho i'"
\ /
2-methy1-1-(tetrahydro-2H-pyran-4- ro, __________ )-----1 y1)-1H-pyrrolo[2,3-131pyridine-3- N 261 carboxylic acid HO I ¨
'I

Name Structure rrt/z ( )-2-methyl-1-(1-(2-oxopyridin-Q.
1(2H)-Apropan-2-y1)-1H- ,,:r.." .

pyrrolo[2,3-b]pyridine-3-carboxylic acid \ r \
( )-2-methy1-1-(1-rfs a Ale (methylsulfonyl)propan-2-y1)-1H- N,r-N

pyrrolo[2,3-b]pyridine-3-carboxylic HO.,TrAtil acid 0 \ 1 ( )-2-methy1-1-(1-(pyr idine-2-yloxy)propan-2-y1)-1H-pytTolo [2,3- s'se 312 HoyLL.6...N
b]pyridine-3-carboxylic acid \ r os 0 ..g....
2-methy1-1-(3-(methylsulfonyl)butan->¨. 310 2-yI)-1H-indole-3-carboxylic acid 1-cyclopenty1-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid __ .....
N
( )-1-(1-cyanopropan-2-y1)-2-methy1-1H-pyrrolor2,3-blpyridine-3- N 244 carboxylic acid HO
........ ....................._ .......
( )-1-(4-amino-4-oxobutan-2-y1)-2- CONH-methyl-1. H-pyrrolo[2,3-b]pyridine-3- HO)r t.Y,..6. )1 I/ 26/
carboxylic acid 0 ---.
( )-2-methyl-1-(1-(1-methy1-2-oxo-) ,,, N-1,2-dihydropyridin-4-yDethyl)-1H- `...,...--N, 312 L "\ `---pyrrolo[2,3-b]pyridine-3-carboxylic HO ---N
acid '10( --L) 1-(3-hydroxy-3-methylbutan-2-y1)-2- Nohi methyl-1H-pyrrolo[2,3-b]pyridine-3- HO i 263 , N
carboxylic acid I
0 ---.
( )-2-methyl.-1-(1-(piperidin-1- \ 2 yl)propan-2-y1)-1H-pyrrolo [2,3- 302 b]pyri di ne-3-carboxyl ic acid 140,*
/ N
i 0 -.-Name Structure rniz ( )-1-(4-hydroxybutan-2-y1)-2-methyl-1H-pyrrolo[2,3-13]pyridine-3-Ho 1 N 249 carboxylic acid 1 o ( )-2-methyl- I -(piperidin- I -ylsulfony1)-1H-pyrrolo[2,3- 324 b]pyridine-3-carboxylic acid OH
111)- I -(2-hydroxypropyI)-2-methyl-1 H-pyrrolo[2,3-blpyri dine-3- HO)Q235 carboxylic acid ( )-1-(3-methoxybutan-2-y1)-2-methyl-1H-indole-3-carboxylic acid 262 ( )- 1 -( 1-methoxypropan-2-y1)-2-methy1-6-(tri fluoromethyl)-1H-pyrrolo [2,3-1Apyridine-3-carboxylic HO 1 N 317 acid \*.),¨
( )-2-methyl-1-(4,4,4-trifluoro-3-\cr3 methoxybutan-2-y1)-1H-indole-3- 316 carboxylic acid HO *

OH
( )- 1 -(3-methoxypentan-2-y1)-2-\
methy1-1H-pyrrolo[2,3-b]pyridine-3 N 277 N
carboxylic acid 1-(3-methoxypentan-2-y1)-2-mc thy 1-11-I-i ndole-3-carboxyl ic acid HO / \
s\

WO 2013/120104 PCT1tJS2013/025639 Name Structure rri/z \. 0 1-(1-methoxy-l-phenylpropan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine- \r¨N 325 3-carboxylic acid F10,119:LO\

1003771 Each of the above carboxylic acids was used as starting material in Step 4 of Example 36 in the synthesis of certain compounds of the invention.
1003781 Example 51. Synthesis of tert-butyl 1-(2.3-dihydro-1H-inden-l-v1)-2-methyl-1H-pyrrolo[23-bills ridint-3-carbovk late. The title compound as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[00379] Tert-butyl 2-methyl-1H-pyrrolor2,3-bi pyridine-3-carboxylate:
otµ
t-butylacetoacetate Cut, L-proline NH
0 Cs2CO3, dioxane Br-b _________________________________________ >ro To a 500 tnL round-bottom flask that contains N-acetyl-N-(3-bromopyridin-2-ypacetamide (14.815 g, 57.6 mmol), was added copper(I) iodide (1.098 g, 5.76 mmol), L-proline (1.327 g, 11.53 mmol), cesium carbonate (28.2 g, 86 mmol), then t-butyl acetoacctate (11.47 ml, 69.2 mmol) and dioxane (100 mL). The reaction was vac/purged with N2 3X then fitted with a septum and a N2 inlet and heated overnight at 70 'C. The inorganic solids were removed by filtration over celite and the cake was washed with 100 mL EtO.Ac. This solution was concentrated and the residue was partitioned between 250 mL brine and 250 mL
Et0Ac. The aq.
Layer was further extracted with Et0Ac (2x250 mL) and the combined organic layer was dried over Na2SO4, filtered, concentrated and purified by CC using 1:1 Et0Ac:Hex as eluent to provide (2.7g, 20.2%) of tert-butyl 2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate. LRMS
+ H+) m/z: calc'd 233.28; found 233.1.
[003801 The compounds shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.

PCT1tJS2013/025639 Name Structure nviz tert-butyl 2-methyl-6 NH
-( trifluoromethyl)-1H-pyrrolo [2,3- >ro 301 b]pyridine-3-carboxylate o k ethyl 6-(4-(tert-butoxycarbonyl)piperazin-l-y1)-2- 389 methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate [00381] Tert-butyl 1-(2,3-dihydro-M-inden-1-y1)-2-methyl-111-pyrrolo[2,3-blpyridine-3-ca rboxylate:

NH OH
N _____________________________________ So' N

A. solution of ethyl tert-butyl 2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (100 mg, 0.74 mmol), 2,3-dihydro-IH-inden-1-ol. (176 mg, 0.74 mmol), PPh3 (195 mg, 1.49 mm.ol) was stirred in dry THF (10 mL) at 0 C under a nitrogen atmosphere. To this mixture was added drop-wise DIAD (150 mg , 1.48 mmol) over a period of 5 min, and the reaction was stired at room temperature for 16 hours. The mixture was washed with brine, dried and concentrated to afford the crude product. The crude product was purified by silea gel chromatography (petroleum ether/ethyl acetate = 5:1) to afford the tert-butyl-1-(2,3-dihydro-1H-inden-l-y1)-2-methyl- I H-pyrrol.o[2,3-14yridine-3-carboxylate (150 mg, 60%0.
1003821 The compounds shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure miz o ( )-tert-butyl 1-(sec-buty1)-2-methyl-1H-pyrrolo[2,3-b] pyridine-3- 289 .1 )¨

carboxylate ( )-tert-butyl 2-methyl-I -(tetrahydrofuran-3-y1)-1H- N 303 pyrrolo[2,3-b]pyridine-3-carboxylate I o Name Structure rn/z.
tert-butyl 1-(chroman-4-y1)-2-methyl-1 H -pyrrolo[2,3-bipyri di ne-3- /N 364 carboxylate 0 I,, ( )-tert-butyl 1-(1-cyclopropylethyl)-2-methy1-1H-pyrrolo[2,3-blpyridine- N 301 3-carboxylate ( )-tett-butyl 2-methyl- 1-(1-ci N xt..) phenylpropyl)-1H-pyrrolo[2,3- 351 b]pyridine-3-carboxylate >ro N-N
( )-tert-butyl 1-(1-(1H-pyrazol-1-yl)propan-2-y1)-2-methy1-1H- N 341 pyrrolo[2,3-b]pyridine-3-carboxylate >I 0 tert-butyl 2-methyl-I -(tetrahydro-2H- r pyran-4-yI)-1H-pyrrolo [2,3- 317 b]pyridine-3-carboxylate 1 _______________________________________________ >r0-1-ONNµ
tert-butyl 1-cyclopenty1-2-methyl- 9 1 H-pyrrolo[2,3-b]pyri dine-3- N 301 carboxyl.ate W-tert-butyl 2-methy1-1-(1-(piperidin-l-yl)propan-2-y1)-1H- 358 pyrrolo[2,3-b]pyridine-3-carboxylate /
[003831 Each of the above alkyl carboxyl.ates was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[003841 Example 52. Synthesis of tert-butvl 2-methy1-143-(methvithio)butan-2-y1)-1H-indole-3-carboxylate. The title compound was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[003851 Step 1: tert-butyl 2-methyl-1(3-oxobutan-2-y1)-1H-indole-3-earboxylate:

\HP
Nr-NH
s tBuoyk.,-3\
KliCs2CO3/
0 CH3CN/reflux To a solution of tert-butyl 2-methyl-1H-indole-3-carboxylate (2g, 8.65mmol) and 3-chlorobutan-2-one (1.1g, 10.38nun01) in acetonitrile (18mL) were added potassium carbonate (3.2g, 25.8mmo1) and potassium iodide (1.4g, 8.65mmo1). The reaction mixture was stirred at 90 C;
overnight. To the reaction mixture was added water (20mL). The aqueous layer was extracted with ethyl acetate (50mL x4). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by silica gel column (petroleum ether/ethyl acetate 100:1 to 80:1) to give tert-butyl 2-methyl- I -(3-oxobutan-2-y1)-1H-indole-3-carboxylate (600mg, yield: 23%) as a yellow oil. LRMS (M+H+) m/z: calcd 301.17; found 302.
1003861 Step 2: tert-butyl 1-(3-hydroxybutan-2-y)-2-methy1-1111-indole-3-carboxylate:

N NaBH4/THF/rt __________________________ so.
tBuOy-Loi To a mixture of tert-butyl 2-methyl-I -(3-oxobutan-2-y1)-11-l-indole-3-carboxylate (120mg, 0.39mmo1) in tetrahydrofuran (3 mL) was added sodium borohydride (79mg, 2.08mmo1). The reaction mixture was stirred at room temperature for 3 hours. The reaction mixture was quenched by adding water (3m1) and extracted with ethyl acetate (20mi., x4). The combined organic layers was dried over sodium sulfate and concentrate to afford tert-butyl 1-(3-hydroxybutan-2-y1)-2-methyl-1H-indole-3-carboxylate (110mg, yield: 93%) as a yellow oil. LRMS (M-1-11+) m/z:
calcd 303.17; found 304.
1003871 Step 3: tert-butyl 2-metby1-1-(3-(tosyloxy)butan-2-y1)-1H-indolle-3-carboxyllate:
OH v.../OTs TsCI
N

1BuO tlEttiO
To a mixture of tert-butyl 2-methyl-1-(3-oxobutan-2-y1)-1H-indole-3-carboxylate (1.1 g, 3.6mmo1) in dichloromethane (4mL) were added 4-methylbenzene-l-sulfonyl chloride (1.3g, 7.2mmol), and 1,4-diazabicyclo[2.2.2]octane (1.2g,10.8mmo1) at 0 C. The reaction mixture was stirred at 0 C for 3 hours. The reaction mixture was added to water (30mL).The aqueous layers was extracted with dichloromethane (10mL x3), The combined organic layers was dried over sodium sulfate and concentrate to afford tert-butyl 2-methy1-1-(3-(tosyloxy)butan-2-y1)-1H-indole-3-carboxylate (1.6g, yield:96 M as a yellow oil. LRMS (M+H+) m/z: calcd 457.19;
found 458.
[003881 Step 4: tert-butyl 2-methyl-1-(3-(methyithio)butan-2-y1)-1H-indole-3-ca rboxylate:
_jars ;4' \
Na tBuo ti3u0 / \

A mixture of compound tert-butyl 2-methyl-1-(3-(tosyloxy)butan-2-y1)-1H-indole-3-carboxylate (200 mg, 0.43 mmol.) in N,N-dimethyl formamide (2mL) was added sodium methanethiolate (994mg, 1.44mmol). The reaction mixture was stirred at 110 C for 4 hours. The reaction was quenched by adding water (6mL). The aqueous layer was extracted with ethyl acetate (30mLx3), and the organic layer was concentrated. The crude product was purified by preparative TLC
(elute: petroleum/ ethyl acetate:5:1) to give product tert-butyl 2-methyl.-1-(3-(methylthio)butan-2-y1)-1H-indole-3-carboxylate (100mg, yield :68%). LRMS (M+H+) m/z: calc'd 333.18, found 334.
1.003891 Step 5: tert-butyl 2-methyl-1-(3-(rnethylthio)butan-2-y1)-1H-indole-3-carboxylate:

\
N" Oxone tu0 q3u0 1 To a solution of compound (4-methoxy-6-methylpyridin-3-yl)methanamine (100mg, 0.3mmo1) in dichloromethane(3mL) and water(3mL) was added oxone (3.7g, 6mm01). The reaction mixture was stirred at room. temperature for 48 hours. The reaction. was quenched by adding water (20mL). The aqueous layer was extracted with dichloromethane (50mLx3) and organic layer was concentrated. The crude product was purified by preparative TLC
(elute: petroleum ether: ethyl acetate 3:1) to give compound tert-butyl 2-methy1-1-(3-(methylsulfonyl)butan-2-y1)-1H-indole-3-carboxylate (30mg, yield: 27.5%).as a yellow solid. LRMS (M+I-1 ) nilz: calc'd 365.17, found 366.
[003901 Example 53. Synthesis of ( R or S)-Ethyl 2-methy1-141-(tetrahydro-2H-pyran-4-yfleth v1)-1 H-PN rrolor2,3-blf)V rid i e-3-ea rboxylate. The title compound was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[003911 Step 1: (R or S)-N-((4-Methoxy-6-methyl-2-oxo-1,2-d d ropy ridin-3-yl)methyl)-2-methyl-1-(1-(tetrahydro-2111-pyran-4-yl)ethyl)-1H-pyrrolo 2,3-b py ri d n e-3-carboxamide:
Cs*

N F Cs+ 0 Os-ry, 0õ) To a re-sealable vial containing 2-fluoro-3-iodopyridine (2.14 g, 9.60 mmol), (S)- or (R)-1-(tetrahydro-2H-pyran-4-ypethanaminium chloride (1.2175 g, 7.35 mmol) and cesium carbonate (7.54 g, 23.14 mmol) was added DMAc (10 mL). The vial was subsequently sealed and placed in a 125 C bath. After stirring at 125 C for 48 h, the reaction mixture was cooled to room temperature and partitioned between Et0Ac with water. The aqueous layer was extracted with Et0Ac (3x). The combined organic layers were washed with a minimal amount of water (2x), dried over Na2SO4, and concentrated to give a heterogeneous brown oil. The oil was diluted with Et0Ac/Hexanes (1:3) and filtered. The solids were washed with Et0Ac:Hexanes (1:3) and the filtrate was concentrated to give a brown oil. The resultant oil was purified on a Biotage system (40 g, gradient elution 2% Et0Ac : 98% Hexanes to 10% Et0Ac : 90% Hexanes, then isocratic 10% Et0Ac : 90% Hexanes). The product (S)- or (R)-3-iodo-N-(1-(tetrahydro-2H-pyran-4-ypethyl)pyridin-2-amine (0.84 g, 2.53 mrnol, 34.4 % yield) was isolated as a clear colorless oil.
LRMS (M +144) m/z: calcd 333.0; found 333.
[003921 The compounds shown in the following table were prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure rn/z WO 2013/120104 PCT1tJS2013/025639 (S)- 3-iodo-N-(1- N NH
p5 phenylethyl)pyridin-2-amine [003931 Step 2: (R or S)-Ethyl 2-methy1-1-(1-(tetrahydro-2H-pyran-4-yl)ethyl)-pyrrolo12,3-bipyridine-3-carboxylate:
OH Am: Cs' nal 0,tr0- 0 Cul Cs-0 Nr. NH +
N N
OE t 0)444' .A re-sealable vial containing (S)- or (R)-3-iodo-N-(1-(tetrahydro-2H-pyran-4-yl)ethyppyridin-2-amine (0.333 g, 1.002 mmol), copper(I) iodide (0.0084 g, 0.044 mmol.), biphenyl-2-ol (0.018 g, 0.106 mmol), and cesium carbonate (0.672 g, 2.062 mm.ol) was diluted with THF
(3.5 mi.). To the orange mixture was added ethyl 3-hydroxybut-2-enoate (0.25 mi., 1.977 mmol). The resultant blue-green contents were evacuated and purged with N2 (g) (3x). The vial was subsequently sealed and heated to 100 'C. After 24 h, the reaction mixture was cooled to room temperature and filtered over a pad of Celite. The filter pad was washed with Et0Ac (3x) and the filtrate was concentrated to give a thick brown oil. The resultant oil was purified on a Biotage system (50 g, gradient elution 2% Et0Ac : 98% Hexanes to 15% Et0Ac : 85%
Hexanes, then 15% Et0Ac : 85% Hexanes). The product (S)- or (R)-ethyl 2-methy1-1-(1-(tetrahydro-2H-pyran-4-ypethyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.257 g, 0.812 mmol, 81.0 % yield) was isolated as a white foam. LRMS (M H.'") nilz: calcd 317.2; found 317.
[00394j The compound shown in the following table was prepared according to the general procedure outlined above using the appropriate starting materials and modifications.
Name Structure miz (S)- ethyl 1 2-methyl- 1-(1-phenylethyl)-10.
1H-pyrrolo[2,3-b]pyridine-3- 309 / N
carboxylate 0 1003951 This compound was also used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
1003961 Example 54, Synthesis of N-04-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-11)methvb-2-methy-1- 1-(143-(methvicarballiOl'OphenvOetliv1)-1H-pyrroloi2.3-bipyridine-3-earboxamide (Compound 263).
[003971 Step 1: 3-(1-(3-(((4-methoxy-6-methy1-2-oxo-1,2-diki, dropyridin -3 -yljmethyl)earbamoy1)-2-methyl-1H-pyrrolo[2,3-blpyridin-l-yljethyljbenzoic acid:
CN
COON
/
(1)1 Li0H, H20, THF, MOH (I) / -7\ reflux /

Lithium hydroxide anhydrate (10.51 mg, 0.439 mmoD in water (5 mL) was added to 1-0 -(3-cyanophenyDethyl)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyrid in-3-yDmethyl)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (20 mg, 0.044 nunol) in tetrahydrofuran (2 mL) and methanol (3 mL) and the resultant mixture was stirred at 100 C for 12 hours.
The mixture was evaporated, added with water (1 mi.), acidified with aqueous hydrochloric acid (1M) to pH = 2.
The precipitate solid was filtered and dried to obtain the title compound (20 mg, yield:96%).
[003981 Step 2: Methyl 3-(1-(3-(04-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methypcarbamoyp-2-methyl-1H-pyrrolo[2,3-b1pyridin-l-ypethyl)benzoate:
c-o COON
112304, Me0H H N
I reflux HN N N
o To a solution of 3-(1-(304-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yDmethyl)carbamoy1)-2-methyl-IH-pyrrolo[2,3-b]pyridin- 1 -yDethyDbenzoic acid (20 mg, 0.042 mmol) in Me0H (6 mL) was added 2-3 drops of sulfuric acid. The mixture was stirred at 70 C
for 1 hour. The solvent was evaporated. The residue was dissolved in water (3 mL), quenched by saturated NaliCO3 solution, extracted with EA (10x3). The combined organic phase was dried by sodium sulfate, and then filtered. The filtrate was concentrated to give the title compound (20 mg, yield: 97%).
100399) Step 3: N44-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1.-(3-(methylearbamoyl)phenyl)ethyl)-1H-pyrrolo[2,3-blpyridine-3-carboxamide (Compound 263):

dI.
\-N
Et0H H
HN HN, 7-N

Methyl 3-(1-(3-(((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)earbamoy1)-2-methyl-1H-pyrrolo[2,3-blpyridin-l-y1)ethyl)benzoate (20 mg, 0.041mmot) was added to 30%
methylamine ethanol solution(10 mL). The mixture was stirred under 50 Psi at 100 C for 12 hours in a 100 mL of sealed tube. The solvent was evaporated and the residue was purified by preparative-HPLC (Instrument: Gilson GX281 Column: ASB C18 150*25nun *5um Mobile phase A: water with 0.05% ammonia solution Mobile phase B: MeCN Column temperature:
30 C Gradient: 17-47% B 15 min) to afford the title compound (17.4 mg, yield:
30%). LRMS
(M m/z:
calcd 488.22; found 488Ø 111 NMR (400 MHz, chloroform-d) 6 ppm 2.01 (d, J=7.28 Hz, 3 H) 2.26 (s, 3 4) 2.50 (s, 3 H) 2.96 (d, .1=4.63 Hz, 3 H) 3.91 (s, 3 H) 4.64 (d, .1=5.51 Hz, 2 H) 5.95 (s, 1 H) 6.14 (br. S., 1 H) 6.55 (d, J=8.16 Hz, 1 H) 7.05 ---7.13 (m, 1 H) 7.33 (t, J=8.05 Hz, 1 H) 7.51 -- 7.59 (m, 1 H) 7.60-- 7.64 (m, 1 H) 8.24 (d, J=5.51 Hz, 2 H).
[004001 Example 55. Synthesis of tert-butvl 3-111rox '-3-methvlbutan-2 1 -2-methvl-M-pyrrolo12.3-nlovridiae-3-carboxviate. The title compound was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
Nie4AgEir -N OH

\ 0 d k 0 THF, -78 0 0 To a solution of tert-butyl 2-mety1-1-(3-oxobutan-2-y1) -1H pyrrolo [2,3-b]
pyridine-3-carboxylate (0.6 g, 2 mmol ) in THF (10 mL) were added CH3MgBr (2 mL, 6 mmol ) at -78 C. The mixture was stirred at -78 C for 3 hr. Water (4 inL) was added and the mixture was extracted by ethyl acetate (30 mL*3). The organic layer was washed with brine and dried over sodium sulfate. The crude product was concentrated and purified by prc-TLC
(eluted: petroleum ether/ethyl acetate = 4/1) to give ter- butyl 1-(3- hydroxy-3-me thy I bu ta n - 2y1)-2-me thy I- 1 H-py rrolo[2,3-b]pyridinc-3-carboxylate (300 mg, 49%).
1004011 Example 56. Synthesis of ( )-tert-butyl 141-(methoxv(methyl)amino)-1-oxopropan-2-y1)-2-methyl-1H-pyrrolo12.3-blnyridine-3-carboxylate. The title compound was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention, 1004021 Step 1: (1)-tert-butyl 1-(1-methoxy-1-oxopropan-2-yI)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-earboxylate:
\ /Jo methyl 2-bromopropanoato , NH
Cs2CO3, DMF
I / /
TM
To a pyrex vial was added tert-butyl 2-methyl- I H-pyrrolo[2,3-b]pyridinc-3-carboxylate (2.491 g, 10.72 mmol) and cesium carbonate (4.54 g, 13.94 nunol). The atmosphere in the vial was vac/purged 3X with N2 then DMF (24 mL) and methyl 2-bromopropanoate (2.393 ml, 21.45 mmol) were added. The reaction was mixed at ambient temperature overnight. The reaction was poured into half-saturated brine and extracted with Et0Ac. The combined organic layer was washed IX each with water then brine, dried over Na2SO4, filtered, deposited onto silica gel and purified by CC (Biotage, 100g column) using 25% Et0Ac in Hex (6 CV) then 50%
(6 CV) as dant to provide ( )-tert-butyl 1-(1-methoxy-l-oxopropan-2-y1)-2-methyl-11-I-pyrrolo[2,3-b]py ridine-3-carboxy late (2.132g, 64%). LAMS (M + H.) ne/z: calcd 319.37;
found 319.2.
1004031 The compound shown in the following table was prepared according to the general procedure outlined in Step I of this example using the appropriate starting materials and modifications.
Name Structure rniz tert-butyl 2-methy1-1-(1-oxo-1-phenylpropan-2-y1)-111-pyrrolo [2.3- N 365 b]pyridine-3-carboxylate -to I / r.µ1 [004041 Step 2: ( )-tert-butyl 1-(1-(methoxy(methyl)amino)-1-oxop rap a n-2-y1)-2-methy1-1H-pyrrol o [ 2 .3-b I pyridine-3-ca rbax-ylate:
\ IF \ iri) 0 1 N)-4(No¨ N,O-dirnethylhypdrmoxygniTiFinFe hydrochloride >r i , sy.....t.) 6 11- 5 rs'rrck ____________________________________________ to >ro I / rsiil 0 To a 1(0 mL round-bottom flask was added (+)-ten-butyl 1-(1-methoxy-l-oxopropan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.958 g, 3.01 mmol) and N,0-dimethylhydroxylamine hydrochloride (0.440 g, 4.51 mmol). The flask was vac/purged 3X
with N2 then THF (17 mL) were added and the reaction was cooled to -40 C.
isopropylmagnesium chloride (4.51 ml, 9.03 mmol) was added dropwise and the reaction was mixed at that temperature for lb then warmed to 0 C for I h then quenched with IN HCI, extracted with Et0Ac. The org layer was washed with water then brine, dried over Na2SO4, filtered, concentrated, deposited onto silica gel with aid of DCM, and purified by CC using 25% Et0Ac in Hex as eluent to provide ( )-tert-butyl 1-(1-(methoxy(methypamino)-1-oxopropan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.630g, 60.3%). LRMS
(M + Fr) mtz: calcd 348.41; found 348.2.
1004051 Step 3: ( )-tert-butyl 2-methy1-1-(3-oxobutan-2-y1)-1H-pyrrolo[2,3-bipyridine-3-earboxylate:
\ 69 MeMyCl. THF 10 µ..)e. ...iXON)-41\
1 N µ I N
/
o - 1 o In a 50 mL round-bottom flask, under an atmosphere of N2, was added ( )-tert-butyl 1-(1-(methoxy(methyl)amino)-1-oxopropan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.630 g, 1.813 mmol) and THF (30 mL). The solution was cooled to 0 C then methylmagnesium bromide (2.59 ml, 3.63 mmol) was added and the reaction was slowly warmed to ambient temperature while being monitored by LCMS. The reaction was done in 3h at ambient temperature. Quenched with 50 ml., 1N HC1, 50 mL brine, extracted with Et0Ac, dried over Na2SO4, filtered, concentrated to provide ( )-tert-butyl 2-methy1-1-(3-oxobutan-2-yI)-1H-pyrrolo[2,3-b]pyridine-3-carboxylate, which was used directly in the following reaction. LRMS (M + m/z: calcd 303.37; found 303.1.
1004061 Examle 57. Synthesis of ( )-N-((41-methox.% -6-meth -1-2-o x 0-1 ,2-d 111%1-Irony rid in-3-vinnethvI)-1-(3-(2-nlethoxyethov0hutan-2-.% l)-2-meth .% 1- 1 11-0-4. rroloi 2,3-h pvridi ne-3-carboxamide 2.2.2-trilluoroacetate (Compound 297), 1004071 Step 1: ( )-tert-buty1-1-(3-(2-met hoxyethox-y)butan-2-y1)-2-methy1-1H-pyrrolo12õ3-b ipyridine-3-earboxylate:
OH
1-bromo-2-methoxyethane , DMF ,N
_________________________ ar >ro.Irke)0 i /2\
Sodium. hydride (0.026 g, 0.657 mmol) and W-tert-butyl 1-(3-hydroxybutan-2-y1)-2-m.ethyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.1 g, 0.329 mmol.) were added to a 25 ml, rbf and the atmosphere was purged with. N2. DMF (5 mL) was added and the reaction was heated at 40 C for lh. The reaction was cooled to ambient temperature and 1-bromo-2-metboxyethane (0.062 ml, 0.657 mmol) was added. The reaction was heated at 90 C for 3d.
LCMS showed ¨60% conversion. The reaction was poured into half-saturated brine, extracted with Et0Ac, the org layer was washed with half-saturated brine, brine, filtered, concentrated and loaded onto a column with aid of DCM. 2X12g column, the column was then treated with 2CV of Hex then 20% EtA0c in Hex to elute (0-tat-butyl 1-(3-(2-methoxyethoxy)butan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (44.4mg, 37.3%). LRMS (M + 111) m/z:
calcd 363.46; found 363.1.
[004081 The carboxylate shown in the following table was prepared according to the general procedure outlined in Step 1 of this example using the appropriate starting materials and modifications.

Name Structure i ( )-tert-butyl 1-(3-ethoxybutan-2-yI)- r¨s\

2-methyl-I H-indole-3-carboxylate 0 1114-Ir 1004091 Step 2: ( )-1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl-1H-pyrrolo[2,3-pyridine-3-carboxylic acid:
r-d TMSOTI, TEA. DOA
\

In a 50 mL round-bottom flask, ( )-tert-butyl 1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (0.0444 g, 0.122 mmol) was added and the atmosphere was vac/purged 3X with N2, then diluted with DCM (10 mL). TEA
(0.026 ml, 0.184 mmol) added and the reaction was cooled to 0 C followed by the addition of TMS-Otf (0.033 ml, 0.184 mmol). The cold bath was removed and the reaction was mixed at ambient temperature for 1h. LCMS showed complete conversion to the carboxylic acid and the reaction was quenched with 50 mL of 1:1 1N HC1 and brine. The aq. Layer was extracted 3x with Et0Ac. The combined org layer was dried over Na2SO4, filtered, concentrated to provide crude )-i( 43-(2-methoxyethoxy)butan-2-y1)-2-methyl- I H-pyrrolo [2,3-bipyri dine-3-carboxylic acid (38mg, 100%), which was used directly in the following reaction. LRMS (M
+ H+) m/z: calcd 307.36; found 307.1.
[004101 The carboxylic acids shown in the following table were prepared according to the general procedure outlined in Step 2 of this example using the appropriate starting materials and modifications.

Name Structure rniz ( )-1-(3-methoxy-3-methylbutan-2-y1)-2-methy1-1H-indole-3-carboxylic 276 acid HO iiiat ( )-11-(3-ethoxybutan-2-y1)-2-methyl-1H-indole-3-carboxylic acid t:t 11111 1004111 Step 3: ( )-1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl- 11-1-pyrrolo12,3-blpyridine-3-carbonyl chloride:
of o--r (COC-02, cat OW
DC3,1 /
________________________ Itk / HON
To To a solution of ( )-1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid (38 mg, 0.124 mmol) in DCM (5 mL, dry at 0 C) was added I
drop of DMF and oxalyl chloride (109 ul, 1.240 mmol). The reaction was monitored by LCMS and upon completion (conversion to methyl ester by quenching an aliquot with Me0H), the volatiles removed, concentrated lx with toluene to provide (-1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl-1H-pyrrolo[2,3-bipyridine-3-carbonyl chloride (40mg, 100%), which was used directly in the following reaction. Methyl ester expected in LRMS (M + Fr) m/z: calcd 321.17; found 321.2.
1004121 Step 4: ( )-N4(4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-1-(3-(2-methoryethoxy)butan-2-yl)-2-methyl-1H-pyrrolo[2,3-bipyridine-3-carboxamide 2,2,2-trifluoroacetate (Compound 297):
o-r4 mi" miA*
TEA, THF C!) HO' F
CI FIN -A,1 o '-To a solution of ( )-1-(3-(2-methoxyethoxy)butan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carbonyl chloride (40.3 mg, 0.124 mmol) and 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(1H)-one hydrochloride (35.5 mg, 0.174 mmol) in THF (5 mL) at 0 C was added TEA (41.5 p.1, 0.298 mmol). The cold bath was removed; the reaction was allowed to stir at ambient temperature and was monitored by LCMS. The reaction was judged to be complete in 3h.
The crude reaction was concentrated then dissolved in 1mi, DMF, 1.5mL Me0H, lmL H20, sonicated, filtered through a PTFE screen and purified by reverse-phase HPLC
using a H20:MeCN:TFA 5-95% 7 min. gradient. The pure product fractions were lyophilized to provide the title compound (16mg, 22.6%) white solid. LRMS (M + 1-1') m/z:
calcd 457.54;
found 457.2. NMR
(400MHz., DMSO-d6) 8 = 11.94 - 11.74 (m, 1 H), 8.19 (dd, J = 1.6, 4.7 Hz, 1 H), 8.10 (dd, J= 1.4, 7.9 Hz, 1 H), 7.97 (br. S., 1 11), 7.10 (dd, J
= 4.7, 7.8 Hz, 1 H), 6.23 (s, 1 H), 4.33 (br. S., 4 II), 3.89 - 3.80 (m, 3 H), 3.27 - 3.16 (m, 1 H), 2.91 - 2.86 (m, 3 H), 2.84 (s, 4 H), 2.67 (s, 3 H), 2.22 (s, 3 H), 1.60 (d, J= 6.7 Hz, 3 H), 1.23 - 1.16 (m, 3 H).
[00413] Example 58. Synthesis of 4-(1-methoxvpropan-2-0-5-methyl-4H-pyrrolo12,3-dIthiazole-6-carboxylic acid. The title compound was used as starting material in Step 4 of Example 36 in the synthesis of certain compounds of the invention.
[00414] Step 1: (Z)-ethyl 2-azido-3-(thiazol-5-yl)acrylate:
Na To a solution of thiazole-5-carbaldehyde (20 g, 176.77 mmol) and ethyl 2-azidoacetate (91.2 g, 707.1 mmol) in anhydrous ethanol (100 mL) was added sodium (16.26 g, 707.1 mmol) dissolved in anhydrous ethanol (1000 mL) dropwise between -10 C and 0 C. After the addition, the mixture was stirred below 0 C for 4 hours, and then warm to the ambient temperature and allowed to stir overnight. The reaction mixture was washed with saturated ammonium chloride, extracted with acetic ester (500mL x 3), the combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 20:1-010:1) to afford the title compound (9 g, 23%) as an yellow solid. LCMS (M + 11-) miz: calcd. 224.04, found 224.9.

1004151 Step 2: Ethyl 4H-pyrrolo12,3-d1thiazole-5-carboxylate:
rd,433131( cH3 N N
cisHo (Z)-ethyl 2-azido-3-(thiazol-5-yl)acrylate (9 g, 40.18 mmol) was dissolved in anhydrous toluene(100 mL), and the resulting reaction was allowed to stir for 2 hours at 120 C. The reaction mixture was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 15:1---*10:1---,8:1) to give the title compound (5g, 63.5%) as an yellow solid. LCMS (M + Fr) calcd. 196.03, found 196.9.
1004161 Step 3: (4H-pyrrolo[2,3-dithiazol-5-yl)methanol:
N N 0 ,N OH
s ( + LIAIH4 _______ 0¨\\
To a solution of ethyl 411-pyrrolo[2,3-d]thiazole-5-carboxylate (5g, 25.48 mmol) and in anhydrous tetrahydrofuran (100 mL) was added lithium aluminum hydride (4.34 g, 127.5 mmol) dissolved in anhydrous tetrahydrofuran (50 mL) dropwise between -10 C and 0 C.
After the addition, the mixture was stirred below 0 C for 1 hours, and then warm to the ambient temperature and allowed to stir for 3 hours at room temperature. The reaction mixture was cooled down to 0 C, quenched by 10 mL water, and then 10 mL 4N sodium hydroxide. The resulting white precipitate was filtered off, washed with acetic ester. The filtrate was dried by anhydrous sodium sulphate. The solvent was removed to afford the crude product (3.5 g, 89%).
The crude product was used directly in the next step. LCMS (M + m/z:
calcd. 154.02, found 154.9.
1004171 Step 4: 5-methyl-411-pyrrolo[2,3-(11thiazole:
N N OH N N
Ept3cSmiH, rTtFA
To a solution of (4H-pyrrolo[2,3-d]thiazol-5-yl)methanol (3.5 g, 22.7 mmol) in anhydrous dichlorometha.ne (50 mL) was added triethylsilane (5.23 g, 45.4 mmol) dropwise below 0 C, trifiuoroacetic acid (5.18 g, 45.4 mmol) was added dropwise followed below 0 C. The resulting reaction system was warm to the ambient temperature and allowed to stir for 2 hours at room temperature. The reaction mixture was poured into saturated sodium bicarbonate, extracted with dichloromethane (50mL x 3), the combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether! acetic ester 15:1¨*8:1) to give the title compound (2.5 g, 80%) as a yellow solid. LCMS (M + m/z: calc'd. 138.03, found 138.9.
[00418] Step 5: 6-bromo-5-methyl-4H-pyrrolo12,3-dithiazole:
N N
N N NBS "?=¨=
S' MOH
Br To a solution of 5-methyl-4H-pyrrolo[2,3-d]thiazole (2.5 g, 18.09 mmol) in acetic acid (50 inL) was added 1-bromopyrrolidine-2,5-dione (3 g, 18.10 mmol) slowly below 0 C, and then warm to the ambient temperature and allowed to stir for 2 hours at room temperature.
The reaction mixture was concentrated, and acetic acid was removed under reduced pressure.
The pH was adjusted to around 7 by progressively adding saturated sodium bicarbonate below 0 C, extracted with acetic ester (50 mI, x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 25:1-415:1¨*10:1) to give the title compound (2.5 g, 64%) as an yellow solid. LCMS (M + 114) m/z: cale'd. 215.94, found 219.2.
[0041.9] Step 6: 6-b rom 0-441-methoxypropan-2-y1)-5-methyl-4H-pyrrolo [2,341]
thiazole:
OMs N N
DMF
Br Br To a solution of 6-bromo-5-methy1-411-pyrrolo[2,3-d]thiazole (2.5 g, 11.52 mmol) in anhydrous N,N-dimethylformamide (20 mL) was added 1-methoxypropan-2-y1 methanesulfonate (3.87 g, 23.03 mmol), and potassium carbonate (3.18 g, 23.03 mmol) followed. The resulting reaction mixture was heated to 70 C, and allowed to stir over night at 70 C. The reaction mixture was washed with water and brine, extracted with acetic ester (30 mL X 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether /
acetic ester 30:1 .. 20:1-310:1) to the title compound (2.0 g, 60%) as orange oil. LCMS (M
+ H ) nilz:
calc'd. 287.99, found 290.9.

1004201 Step 7: 4-(1-methoxypropan-2-y1)-5-methy1-4H-pyrrolo [ 2,3-d th azole-6-carbonitrile:
N N
CuCN
DMF
Br CN
To a solution of 6-bromo-4-(1-methoxypropan-2-y1)-5-methyl-4H-pyrrolo[2,3-dithiazole (2.0 g, 6.92 mmol) in anhydrous N,N-dimethylformamide (15 mL) was added cyano copper (1.24 g,
13.84 mmol). The resulting reaction mixture was heated to 150 C and allowed to stir for 2 hours at 150 C. The reaction mixture was washed with water and brine, and extracted with acetic ester (30 triL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 30:1¨)20:1¨>10:1) to give the title compound (1.0 g, 62%) as yellow oil. LCMS (M + Fr) m/z: calc'd. 235.08, found 236Ø
[004211 Step 8: 441-methoxypropan-2-y1)-5-tnethyl-4H-pyrrolo[2,3-dIthiazole-6-carboxylic acid:
NaOH
_______________________ _ MeOHITHFIH20 ON Reflux COOH
To a solution of 4-(1-methoxypropan-2-y1)-5-methyl-4H-pyrrolo[2,3-dithiazole-6-carbonitrile (1.0 g, 4.25 mmol) in tetrahydrofuratilmethano1=1:1 (30 mL) was added sodium hydroxide (680 mg, 17 mmol) resolved in 15 mL water. The resulting reaction system was heated to 80 C and allowed to stir for 24 hours at 80 C. The pH was adjusted to around 7 by progressively adding 4N hydrogen chloride below 0 C, solvent and water was removed in reduced pressure to give the title compound (500mg, 46%) as an yellow solid. LCMS (M + 11+) rn/z: calc'd.
254.07, found 254.7.
100422l Example 59. Synthesis of ( )-tert-butyl 1-f3-methoxv-3-rnethylha tan-2- sl)-2-meth l-indole-3-carboxylate. The title compound was used as the starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
1004231 Step 1: ( )-tert-butyl 1-(3-hydroxy-3-methylbutan-2-y1)-2-methy1-111-indole-3-carboxylate:

0 511.1 MeLi, TM;
N
o \

A 50 ml. round bottom flask was charged with ( )-tert-butyl 2-rnethy1-1-(3-oxobutan-2-y1)-1H-indole-3-carboxylate (0.2441 g, 0.810 mmol), vac/purged with N2, diluted with THF (5 mL) and cooled to -78 'C. Methyllithium (0.648 ml, 0.972 nun61) was then added slowly, drop-wise, and the solution was allowed to slowly warm to ambient temperature. The reaction was then quenched with IN HCI, extracted with Et0Ac. The org layer was washed with water then brine, dried over Na2SO4, filtered, concentrated, purified by column chromatography using 10% Et0Ac in Hex (LOCI), 15% (IOCV) to elute ( )-tert-butyl 1-(3-hydroxy-3-methylbutan-2-y1)-2-methy1-1H-indole-3-carboxylate (157.2mg, 61.1%). LRMS (M + fr) m/z: calcd 318.42;
found 318.2.
[004241 Step 2: ( )-tert-butyl 143-methoxy-3-methylbutan-2-y1)-2-methyl4H-indole-3-carboxylate:
NaH, Mel, THF
Nf Tar *
>10 I

To a solution of ( )-tert-butyl 1-(3-hydroxy-3-methylbutan-2-y1)-2-methy1-1H-indole-3-carboxylate (0.2594g. 0.817 mmol) and THF (8 mL), in a 50 mL round bottom flask, cooled to 0 C was added sodium hydride (0.065 g, 1.634 mmol). The reaction was then heated at 45 C for 2h then iodomethane (0.102 ml, 1.634 mmol) was added and the reaction was heated at that temperature overnight. The reaction was quenched with IN HC1 and brine, extracted with Et0Ac, dried over Na2SO4, filtered, concentrated, deposited onto silica gel with aid of DCM, and purified by column chromatography using 5% Et0Ac in Hex as eluent to provide ( )-tert-butyl I -(3-methoxy-3-methylbutan-2-y1)-2-methy1-1H-indole-3-carboxylate (257.8mg, 95%)). LRMS
+ Fr) m/z: calcd 332.45; found 332.2.
[004251 Example 60. Synthesis of (tert-butyl 1-0-methoxybutan-2-v1)-2-methvi-indo1e-3-carboxylate). The title compound was used as the starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[004261 Step 1: (tert-butyl 1-(3-hydroxybutan-2-y1)-2-methy1-1.11-indo1e-3-carboxylate):

Ho N
+ NaBH4 \

0 =
To a solution of tert-butyl 2-methyl-1-(3-oxobutan-2-y1)-1H-indole-3-carboxylate (550 mg, 1.83 mmol) in THF (8 mL) was added NaBH4 (139 mg, 3.66 nunol) at 0 C under N2. The reaction was stirred at 31 C for 20 hrs. The mixture was diluted with ethyl acetate, washed with water and saturated aqueous NaHCO3. The organic layer was concentrated in vacuo to afford the title compound (520 mg) as a yellow oil which was used directly without purification.
[00427j The compound shown in the following table was prepared according to the general procedure outlined in Step 1 of this example using the appropriate starting materials and modifications.
Name Structure mlz tert-butyl 1-(1-hydroxy-l-phenylpropan-2-y1)-2-methy1-1H- N 367 pyrrolo[2,3-b]pyridinc-3-carboxylatc o ¨
1004281 Step 2: (tert-butyll 1-(3-m ethoxybu tan-2-yI)-2-methyl-1H-in dole-3-carboxylate):
HO /
\N. + NaH + Mel To a suspension of tert-butyl 1-(3-hydroxybutan-2-y1)-2-methy1-1H-indole-3-carboxylate (500 mg, 1.65 rnmol) in THF (8 mL) was added sodium hydride (330 mg, 8.25 mmol) at 30 C under N2. The reaction was stirred for 20 min. Then iodomethane (0.4 mL) was added and the reaction was stirred at 60 C for 3 hrs. Then the mixture was diluted with ethyl acetate, washed with water and saturated aqueous NaHCO3. The crude product was purified by column chromatography on silica gel eluted with petroleum ether: ethyl acetate = 80: 1 to afford the title compound as a yellow solid which was used directly (400 mg, 76.8 %).

(004291 The compounds shown in the following table were prepared according to the general procedure outlined in Step 2 of this example using the appropriate starting materials and modifications.
Name Structure ______________________________________________ miz ( )-tert-butyl 2-methy1-1-(4,4,4-trifluoro-3-methoxybutan-2-y1)- I H- i 372 indole-3-carboxylate >r0 _______ ( )-tert-butyl 1-(3-methoxypentan-2-y1)-2-methyl-IH-pyrrolo[2,3- \

b]pyridine-3-carboxylate MN
\0 ( )-tcrt-butyl 1-(3-methoxypentan-2-y1)-2-methyl- I H-indole-3- N 332 carboxylate I
tert-butyl1-(1-methoxy-1- 0 phenylpropan-2-y1)-2-methy1-1H- 381 pyrrolo[2,3-b]pyridine-3-carboxylate [00430] These t-butyl carboxylates were also used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
[00431] Example 61. Synthesis of tert-butvi 2-methvI-144, 4 4-trifitioro-3-hydrowbutan-2-v11-1H-vvrrolo 12. 3-bl pyridine-3-carboxylate. The title compound was used as the starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
1004321 Step 1: 2-(3-(tert-butoxycarbony1)-2-methyl-1H-pyrrolo [2, 3-b]
pyridin-1.-y1) propanoic acid:

o CO¨ + LOH
N" N

To tert-butyl 1-(1-ethoxy-l-oxopropan-2-y1)-2-methy1-1H-pyrrolo [2,3-b) pyridine-3-carboxylate (760mg, 2.29mmol) in CH3OH (5m1) and H20 (5m1) were added LiOH
(548mg, 22.86mm01) at 25 C, the solution was stirred for 3h.After the reaction completed. The solution was concentrated under vacuum and acidified to PH 3 with HC1 (1 M), solid was precipitate out after adjusted. The mixture was extracted with Et0Ac. The organic layer was separated and washed H20 and dried over Na2SO4, and evaporated under reduced pressure to give 6-methy1-7-(1-phenylethyl) pyrrolo [1, 2-b] pyridazine-5-carboxylic acid as a white oil (760mg, yield 100%).
[004331 Step 2: Synthesis of tert-butyl 1-(1-hydroxypropan-2-y1)-2-methyl-1H-pyrrolo (2, 3-131 pyridine-3-carboxyhtte:
o o \\F
i=stiocc;
\
NaBH4
14 N N
H
\ON
To a solution of 6-methy1-7-(1-phenylethyl) pyrrolo [1, 2-b] pyrida7ine-5-carboxylic acid (760mg, 2.5mmol), NMM (505mg, 4.99mmo1) in THF (10 ml) was added i-BuOCCI
(512mg, 3.75mmo1) dropwise at -15 C for 15min. and added NaBH4 (283mg, 7.49mmo1) in portions. The solution was allowed to warm 25 C and stirred for 1h. The reaction mixture was added H20 (0.18m1) dropwise at 0 C for 5 min. The mixture was allowed to warm 25 C and stirred for 1h.
After the reaction completed, the solution was concentrated under vacuum. The solid was partitioned between DCM and 1120. The organic layer was separated and washed TCA(1%) and dried over Na2SO4, and evaporated under reduced pressure to give tert-butyl hydroxypropan-2-y1)-2-methy1-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate as white oil (700mg, yield 97%).

1004341 Step 3: Tert-butyl 2-methyl-1-(1-ox op rop a n-2-y1)-1H-pyrrohif 12, 3-b1 pyridine-3-earboxylate:
0 \/". 0 s. \A-sr 0 DI" \>-**14.-/\--1 To tert-butyl 1-(1-hydroxypropan-2-y1)-2-methy1-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate (380mg, 1.31mmol) in CH2C12(5m1) were added DMP (610.6mg, 1.44mmo1) at 25 C
for 18h.
After the reaction completed, the solution was purified by silica gel and concentrated under reduced pressure to give tert-butyl 2-methy1-1-(l-oxopropan-2-y1)-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate as a white oil (200mg, yield 53%).
[004351 Step 4: Tert-butyl 2-methyl-1-(4, 4, 4-trifluoro-3-hydroxybutan-2-y1)-1H-pyrrolo [2,3-b) pyridine-3-carboxylate:
0, 0 1 .0"4=Kõ, 1 IMS`CF3 TBAF --"1"" Nr "
(pH
.A---F
F
To a solution of tert-butyl 2-methyl-H1 -oxopropan-2-51)-1H-pyrrolo [2, 3-b]
pyridine-3-carboxylate (100mg, 0.35nuno1) in dry THF (2m1) was added a solution of TMS-CF3 (0.02m1, 1M in THF) and the reaction mixture was cooled to 0 C under Ar, Added TBAF
(73.97mg,0.52rnm01), via drop-wise addition and stirred for 30min at 0 C. A
solution of saturated NRIC1 was added and most of the solvent was stripped on the ratary evaporator. The reminder was taken up Et0Ac and 120 and transferred to a separatory funnel.
The mixture was shaken and the organic layer was separated, dried over Na2SO4, and evaporated under reduced pressure to give tert-butyl 2-methyl- l-(4. 4, 4-trifluoro-3-hydroxybutan-2-y1)-1H-pyrrolo [2, 3-b]
pyridine-3-carboxylate as a white crystal (40mg, yield 32%).

1004361 Example 62. Synthesis of 141.-methoxv-2-methvIpropan-2-v1)-2-methy1-1H-indole-3-earbox 1k acid. The title compound was used as the starting material in Step 4 of Example 36 in the synthesis of certain compounds of the invention.
1004371 Step 1: Ethyl 2-(2-methyl-1H-indo1-1-yl)acetate:

Fl N
Cs2CO3 OMP
To a solution of 2-methyl-1H-indole [2,3-d]thiaz,ole (1.0 g, 7.6 mmol) in anhydrous N,N-dimethylformarnide (10 mL) was added ethyl 2-bromoacetate (1.9 g, 11.4 mmol), and cesium carbonate (3.7 g, 11.4 mmol) followed. The resulting reaction mixture was heated to 70 C and allowed to stir for 4 hours at 70 C. The reaction mixture was washed with water and brine, extracted with acetic ester (30 mL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel (eluted with petroleum ether / acetic ester 30:1¨*20:1---410:1) to give ethyl 2(2-methy1-1H-indo1-1-ypacetate (1.5 g, 90%) as a yellow oil. LCMS (M +
Fr) m/z:
calcd. 217.11, found 217.9.
1004381 Step 2: Ethyl 2-meth3.1-2-(2-inetth1-1H-indo1-1-,1)propaimate:

MeliNaH
DiAr To a solution of ethyl 242-methyl-I H-indo1-1-ypacetate (1.4 g, 6.4 mmol) in anhydrous N,N-dimethylformamide (10 mL) was added sodium hydride (1.6 g, 64.4 mmol) at 0 C, then it was allowed to stirred at CPC.After 1 hour, iodomethane (9.1 g, 64.4 mmol) was added dropwise at 0 C. The resulting reaction mixture was allowed to stir for 4 hours at room temperature. The reaction mixture was quenched by adding water (10m1). The pH was adjusted to around 6 by progressively adding 2N hydrogen chloride below 0 C, extracted with acetic ester (50 mL x 3).
The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petroleum ether / acetic ester 50:1-60:1-410:1) to give ethyl 2-methy1-242-methy1-1H-indol-1-yl)propanoate (1.2 g, 76%) as a light yellow oil. LCMS (M + mlz:
calcd. 245.14, found 245.9.

[004391 Step 3: 2-methyl-2-(2-methyl-1111-indol-1 -yl)p rop a n -1-o1:

-.1,1)1¨NH
LAH
/ THF
To a solution of ethyl 2-m.ethyl.-2-(2-methyl.-114-indol-1-y1)propanoate (1.2 g, 4.9 mmol) in anhydrous tetrahydrofuran (10 mL) was added lithium aluminum hydride (1 g, 24.5 mmol.) dissolved in anhydrous tetrahydrofuran (5 mL) dropwise between -10 C and 0 C.
After the addition, the mixture was stirred below 0 C for 1 hour, and then allowed to stir for 3 hours at room temperature. The reaction mixture was cooled down to 0 C, quenched by 10 mL water, and then 10 mL 4N sodium hydroxide. The resulting white precipitate was filtered off, washed with acetic ester. The filtrate was dried by anhydrous sodium sulphate. The solvent was removed to afford the crude product an purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 20:1-410:1¨>5:1) to give 2-methyl-2-(2-methyl-1H-indo1-1-yl)propan-1 -01 (780 mg, 79N as a light green oil. LCMS (M + H m/z: calcd. 202.14, found 202.9.
1004401 Step 4: 1-(1-methoxy-2-methylp fop a n-2-y1)-2-methyl-IH-indole:
OH MoliNaH
N
I
DMF
To a solution of 2-methyl-2-(2-methyl-ift-indol-1-yl)propan- 1 -ol (780 mg, 3.8 mmol) in anhydrous N,N-dimethylfonnamide (10 mL) was added sodium hydride (919 mg, 38.3 mmol) at 0 C. The resultant reaction mixture was allowed to stirred at 0 C for lb.
Iodomethane (5.4 g, 38.3 mmol) was added dropwise at 0 C, And then the resulting reaction mixture was allowed to stir for 3 hours at room temperature. The reaction mixture was quenched with water. The pH was adjusted to around 6 by progressively adding 2N hydro chloride below 0 C, extracted with acetic ester (50 mL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether / acetic ester 10:1-0:1) to give 1-(1-methoxy-2-methylpropan-2-y1)-2-methy1-1H-indole (800 mg, 96%) as a light yellow oil. LCMS (M + H+) m/z:
calcd. 217.15, found 217.9.
1004411 Step 5: 1 -(1 -methoxy-2-methylpropan-2-y1)-2-m ethy1-1 Fl-i ndole-3-ca rbaldehyde:

P0a3 o I / DMF/DCM I

To a solution of 2 ml anhydrous N,N-dimethylformamide dissolved in 80 ml anhydrous dichloromethane was added phosphoryl trichloride (5 g, 32.6 mmol) at 0 C. The resultant reaction mixture was allowed to stirred at 0 C for 3 hours. 1-(1-methoxy-2-methylpropari-2-y1)-2-methy1-1H-indole (800 mg, 3.7 mmol) was added dropwise at 0 C. Then the resulting reaction mixture was allowed to stir for 1 hour at 0 C, and then warmed to the ambient temperature. The reaction mixture was allowed to stir for 24 hours at room temperature. The reaction mixture was quenched with saturated sodium acetate. The pH was adjusted to around 8 by progressively adding 2N sodium hydroxide below 0 C, extracted with dichloromethane (100 mL x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph on silica gel eluted with (petrol ether acetic ester 10:1¨.5:1¨.2:1) to give 1-(l -methoxy-2-methylpropan-2-y1)-2-methy1-1H-indole-3-carbaldehyde (510 mg, 57%) as a light yellow oil. LCMS (M H+) miz: calcd.
245.14, found 245.9.
1004421 Step 6: 1-(1-methoxy-2-methylpropan-2-y1)-2-niethy1-1H-indole-3-carboxylic acid:

Kt,An04 T ?---acetorelti20 .. , ci To a solution of 1-(1-methoxy-2-methylpropan-2-y1)-2-methy1-1H-indole-3-earbaldehydc (50 mg, 0.2 mmol) in acetone (2 mL) and water (2 inL) was added potassium permanganate (95 mg, 0.6 mmol). The resulting reaction mixture was allowed to stir for 3 hours at room temperature.
The reaction mixture was lyophilized directly to give 1-(1-methoxy-2-methylpropan-2-y1)-2-methy1-1H-indole-3-carboxylic acid (50 mg, 94%) as a brown solid. LCMS (M +
miz: calcd.
261.14, found 261.9.

1004431 Example 63. Synthesis of tert-butvl 1-(3-hvdroxypentan-2-vb-2-inetlik1-111-pyrrolo 12, 3-bt pYridine-3-carboxylate. The title compound was used as starting material in Step 3 of Example 36 in the synthesis of certain compounds of the invention.
1004441 Step 1: 2-bromopentan-3-one:
j cuar2 To pentan-3-one (5.0 g, 58.05 mmol) in CHCI3 (25 mi.,) and Et0Ac (25 MI) were added CuBr2 (13.0 g, 58.05 mmol) at 70 C. The solution was stirred at 70 C for! 8h. After the reaction completed, the reaction mixture was cooled to r.t. and filtered through a Celite pad. The filtration was evaporated under vacuum to give 2-bromopentan-3-one as green oil. (8.0 g, yield 84 %) [00445] Step 2: Tert-butyl 2-methy1-1-(3-oxopentan-2-y1)-1H-pyrrolo [2, 3-b]
pyridine-3-carboxylate:
\

0, i `sr r b The solution of ert-butyl 2-methyl-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate (1.0 g, 4.31 mmol), 2-bromopentan-3-one (1.1 g, 6.46mmo1), Cs2CO3(2.8 g, 8.61 mmol), KI
(142.9mg. 0.86 mm.ol) in CH3CN (4m1) were stirred at 70 C for 2h. After the reaction completed, the solution was cooled to r.t. and filtered off. The filtration was evaporated under vacuum. The residue was purified by flash column (Eluent: PE: Et0Ac=5:1) to get tert-butyl 2-methy1-1-(3-oxopentan-2-y1)-1H-pyrrolo [2, 3-1)] pyridine-3-carboxylate as a white oil. (100 mg yield 7 %) [00446] Step 3: Tert-butyl 1-(3-hydroxypentan-2-y1)-2-methy1-111-pyrrolo [2, 3-b]
pyridine-3-carboxylate:

NaBH4 =Ns N
N N

OH
To tert-butyl 2-methyl-1-(3-oxopentan-2-y1)-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate (100 mg, 0.32 annol) in CH3OH (2 ml) were added NaBH4 (35.9 mg, 0.95 mmol) at 0 C.
The mixture was allowed to warm 25 C with stir for 2 h. After the reaction completed, the solution was concentrated under vacuum and the solid was partitioned between Et0Ac and H20.
The organic was separated, dried over Na2SO4 and evaporated under reduced pressure to give tert-butyl 1-(3-hydroxypentan-2-y1)-2-methy1-1H-pyrrolo [2, 3-b] pyridine-3-carboxylate as a white oil. (50 mg yield 50 %).
[004471 Example 64. Synthesis of ( )-N-((4-methoxy-6-methyl-2-ox o- I ,2-di hvdron v rid i n-3.-vi)niethvl)-1-(1-methoxvpropan-2-v1)-2-methyll-6-(pipe I-I-p yrrolof 2,3-b I ovridine-3-carboxamide (Compound at2n.
[00448l Step 1: Tert-butyl 4-(6-aminopy 1)piperazine-1 -carboxy late:
oc H2N N, J
HN

A mixture of 6-chloropyridin-2-amine (10 g, 78 mmol) and tert-butyl piperazine-1-carboxylate (29 g, 156 mmol) was refluxed at 140 C for 3 days. The reaction mixture was purified by silica gel column chromatography (elute: petroleum ether / ethyl acetate = 2:1) to give tert-butyl 4-(6-aminopyridin-2-yl)piperazine-1-carboxylate (6 g, 28%). LCMS (M H) infz:
calcd 278.17;
found 279.
[004491 Step 2: Tert-butyl 4-(6-amino-5-bromopyridin-2-yl)piperazine-1-carboxylate:
r---N-13 c + N¨Br H2N
BrLT

To a solution of tert-butyl 4-(6-aminopyridin-2-yppiperazine- 1 -carboxylate (1.9 g, 8 mmol) in N,N-dimethylformamide (20 ml) was added dropwise 1-bromopyrrolidine-2,5-dione (1.4 g, 8minol) at 0 C. The reaction was allowed to stirred for 4h at rt. The reaction mixture was quenched with water (30 mL), and extracted with acetic ester (30 MI, x 3). The combined organic phase was dried by anhydrous sodium sulphate, and then filtered. The filtrate was concentrated and purified by column chromatograph silica gel (elute: petroleum ether / ethyl acetate = 1:1) to give tert-butyl 4-(6-amino-5-bromopyridin-2-yl)piperazine-1 -carboxylate (0.2 g, 8.2%) as a yellow solid. LCMS (M + calcd. 356.08; found 357.
[004501 Step 3: Tert-butyl 4-(3-(((4-metboxy-6-methy1-2-oxo-1,2-dihydropyrid in-3-yl)methyl)carbamoy1)-1-(1-metboxypropan-2-y1)-2-methy1-1H-pyrrolo[2.3-b]
pyridin-6-yl)piperazine-1-carboxylate:
Jo¨

/
8c)c , -""=-rsiros=-= HATLYTEA/Dc,µ10 N
jµi N HNp.N1-12 I N/Th 0 0 L,N,Boc To a solution of 1-(1-methoxypropan-2-y1)-2-methy1-6-(piperazin-l-y1)-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid (80 mg, 0.19 mmol) in anhydrous dichloromethane (10 mL) was added 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(111)-one (65 mg, 0.38 mmol), 2-(7-Aza-1H-benzotriazole-1-y1)-1,1,3,3-tetrarnethyluronium hexafluorophosphate (145 mg, 0.38 mmol) and triethylamine (96 mg, 0.95 mmol). The mixture was stirred at room temperature for 24 hour.
The reaction mixture was concentrated and used directly in the next step. LRMS
(M+H) m/z:
calcd 582.69; found 584.
[004511 Step 4: (*)-N4(4-metboxy-6-metbyl-2-oxo-1,2-dihydropyridLin-3-y1)methyl)-1.-(1-methoxypropan-2-y11)-2-methy1-6-(piperazio-1-y1)-1H-pyrrolo12,3-bipyridine-3-carboxamide (Compound 295):
HCI
=

HN NPs\
0 0 LIN¨Boc To tert-butyl 4-(3-(((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)carbamoy1)-1-(1-m ethoxypropan-2-y1)-2-methy1-1H-pyrrolo [2,3-14yri din -6-yl)piperazine-l-carboxylate (crude ,used directly from last step). was added saturated hydrochloride solution in methanol (5m1) at 0 C. The reaction mixture was allowed to room temperature and stirred for 2 hours. The reaction mixture was concentrated and purified by preparative HPLC (Mobile phase A : water with 0.05% ammonia solution; Mobile phase B: MeCN; column temperature: 30 C
Gradient:

30-60% B 10 min) to give a N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-(1-methoxypropan-2-y1)-2-methy1-6-(piperazin-l-y1)-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (23 mg, 24.7%, two steps.). LAMS (M -1-H) in/z: cake). 482.26; found 483. NMR
(400 MHz, CDC13) 6: 1.59-1.62 (d, J= 7.2 Hz, 3H), 2.21 (s, 3H), 2.71 (s, 3H), 2.96-3.01 (m, 4H), 3.23 (s, 3H), 3.40-3.43 (m, 4H), 3.81-3.88 (m, 4H), 4.10-4.14 (m., 1H), 4.61-4.63 (d, J
= 5.6 Hz, 2H), 5.91 (s, 1H) , 6.41-6.44 (d, J= 8.8 Hz, 1H), 7.41-7.45 (m, 1H) , 7.90-7.93 (d, J= 8.8 Hz, 1H).
[00452] Example 65. Synthesis of isolated N-((4-methoxy-6-methvl-2-oxo-1õ.2-dihydropyridin-3-vpmethvi)-1.42R or 2S, 3R or 3S)-3-methoxybutan-2-1/1)-2-tnethyl-11-1-pvirroloi2,3-blovridine-3-earboxamide diastereomers (Compounds 261, 266, 267 and 302).
[00453] Step 1: Teri-butyl 2-me1 hy1-1-(3-oxobutan-2-y1)-1H-pyrrolo[2,3-blpyridine-3-carboxylate:
CD.N, + + Cs2CO3 + K\

o To a solution of tert-butyl 2-methy1-1H-pyrrol.o[2,3-b]pyridine-3-earboxylate (5.0 g, 21.53 mm.ol) in CH3CN (50 mL) was added Cs2CO3 (21.0 g, 64.58mmol), potassium iodide (3.57 g, 21.53 mm.o1). The mixture was stirred at 27 C for 30 minutes. Then 3-chlorobutan-2-one (2.75 g, 25.83 mmol) was added and the mixture was stirred at 70 C for 12 hours. The mixture was filtered and the filtrate was concentrated. The residue was purified by column (Elute: Petroleum ether: Ethyl acetate =50:1) to give tert-butyl 2-methyl.-1-(3-oxobutan-2-y1)-1.F1-pyrrolo[2,3-b]pyridine-3-carboxylate as a yellow-green oil.(3.23 g, yield 50 %) LCMS (M
Er) tn/z: calcd 303.37; found 302.9. III NMR (400 MHz, CDC13): a 8.32-8.30 (m, 1.H.), 8.25-8.23 (m, 1.F1), 7.17-7.14 (m, 1H), 5.50-5.44 (m, 1H), 2.71(s, 3H), 1.96 (s, 311), 1.65-1.67 (d,3H), 1.64 (s, 911).
[00454] Step 2: Tert-butyl 1-(3-hydroxybutan-2-y1)-2-methy1-1H-pyrro1o[2,3-b[pyridine-3-carboxylate:
HO/ m <0. + NaBH4 4 memi To the solution of tert-butyl 2-methy1-1-(3-oxobutan-2-y1)-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (3.1 g, 10.25 mmol) in methanol (30 mL) was added sodium borohydride (0.30 g, 8.2 mmol) at 0 C. After 30 minutes, another batch of sodium borohydride (0.30 g, 8.2 mmol) was added at 0 C. After the reaction completed about 2 h later, water (30 ml) was added dropwise very carefully to quench the reaction. The mixture was extracted with CH2C12. The extraction was dried over Na2SO4, filtered and concentrated under vacuum to give tert-butyl 1-(3-hydroxybutan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate as a yellow solid. (3.0 g, yield 96 %) LCMS (M + H-) miz: calcd 305.38; found 304.9. 1H NMR (400 MHz, CDC13): a 8.31-8.29 (m, 1H), 8.13-8.12 (m, 1H), 7.11-7.07 (m, 1H), 4.46-4.43 (m, 1H), 4.12 (m, 1H), 2.73 (s, 3H), 1.58 (s, 9H), 1.51-1.49 (d, 3H), 0.92-0.91 (d, 3H).
[00455] Step 3: Tert-buty1-1-(3-methoxybutan-2-y1)-2-methyl4H-pyrralo[2,3-bipyridine-3-carboxylate:
HO
I _____________________ To dry THF (20 mL) was added NaH (60 % in mineral oil, 2.37 g, 59.14 mmol).
Then the mixture was stirred at 27 C for 20 minutes, then tert-butyl 1-(3-hydroxybutan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylate (3.0 g, 9.86 mmol) was added. The mixture was stirred at 27 C for 1 hour, then added by CH3I (13.99 g, 98.6 mmol). The mixture was stirred for 12 hours at 27 C and then cooled to 0 C. Sat. NH4CI was added and extracted with CH2C12. The extraction was dried over sodium sulfate, filtered and concentrated to give tert-butyl- l -(3-methoxybutan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxylate as a yellow oil. (3.2 g, yield 100%) LCMS (M + H-) rn/z: calcd. 319.41; found 318.9.
[00456] Step 4: 1-(3-methoxybutan-2-y1)-2-methyl4H-pyrrolo[2,3-blpyridine-3-carboxylic acid:

¨0 N
TFA
N.., AI, ; r TFA
-;-;
HO
, \ a To the pre-cooled solution of tert-butyl 1-(3-methoxybutan-2-y1)-2-met41-1H-pyrrolo[2,3-14yridine-3-carboxylate (3.0 g, 9.42 mmol) in CH2C12 (20 mL) was added trifluoroacetic acid (20 mL) dropwise. The solution was stirred at 27 C for 1.5 hours. The solvent was removed under vacuum at 27 C. The residue was used for next step without purified.
LCMS +
m/z: calcd 263.30; found 262.9.
[004571 Step 5: N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-y1)methyl)-1.-(3-methoxybutan-2-y1)-2-methyl-1.11-pyrrolo12,3-blpyridine-3-carboxamide:

OLNµ'14 \ 0 + 112N 11111 + \_N\> I N
TFA
0 # N
\ 0 To a solution of 143-methoxybutan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid (2.4 g, 9.15 mmol) in DMF (30 mL) was added TEA (4.2 g, 41.50 mmol), 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(1H)-one hydrochloride (2.1g, 12.81 mmol) After stirred for 10 minutes at 27 C, the mixture was cooled and added HAM (5.56g, 14.64mmo1). The mixture was stirred at 27 C for 72 hours and 30 % of S.M. remained. Then the mixture was heated at 80 C for 5 hours. The solution was diluted with brine (100 mL) and extracted with CH2C12 (100 mL*3). The extractions were combined and dried over Na2SO4. The solvent was evaporated under vacuum and the residue was purified by flash column (Eluent:
dichloromethane: methanol =95:5) to give N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyri di n-3-y1 )methyl)-1-(3-methoxybutan-2-y1)-2-methy1-111-pyrrolo[2,3-14yridinc-3-carboxamide. (3.6g, yield 95 A) 1004581 Step 6: Separation of N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-y1)methyl)-1-(3-methoxybutan-211)-2-methyl-1H-pyrrolo[2,3-131pyridine-3-ca rb ox anti d e:
Isomers (Compounds 261, 266, 267, and 302):

\O
I )¨

SFC N N NN./ iNilry:.:6 HN I N N HN I /

Method 94 Compound 261 Compound 266 Major Isomer Peak 1 Peak 2 \O \ \
I
SFC

Method 94 Compound 267 Compound 302 Minor Isomer Peak Peak 2 The mixture of isomers from Step 5, N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl )meth y1)-1-(3-methoxybu tan-2-y1)-2-methy1-1H-pyrro lo [2,3-12] pyridine-3-carboxamide was purified by prep-HPLC (Condition: Column: SHIMADZU LC-8A, 250*50mm *10um;
Mobile phase A: water with 0.2 % formic acid; Mobile phase B: McCN; column temperature: 30 C;
Gradient : B in A 10-50 %) to give a major isomer pair (Compound 261 and Compound 266 combined) (1.0 g, purity 98.8%) and a minor isomer pair (Compound 267 and Compound 302 combined) (180 mg, purity 63%). The resulting isomer pairs were individually separated by SFC
(Condition: Column: ChiralpdcmAD 250*30 mm *5 urn; Mobile phase A:
Supercritical CO2;
Mobile phase B: IPA-FM-134120; Gradient : 75:25) to give the following individual single compounds:
1004591 Compound 261, N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-y1)methyl)-1-((2R or 2S, 3R or 3S)-3-methoxybutan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (Major Isomer Pair; Peak 1): ill NMR (400 MHz, CDC13): 8 8.173-8.157 (m, 1H), 8.140-8.116 (m, 111), 7.582-7.555 (m, 11-1), 6.968-6.936 (m. I H), 5.927 (s, 111), 4.707-4.609 (m, 214), 4.348 (s, 114). 3.892 (s, 3H), 2.869 (s, 31-1), 2.788 (s, 311), 2.173 (s, 314), 1.644-1.627 (d, 3H), 1.263-1.249 (d, 311).
1004601 Compound 266, N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yOmethyl)-1-((2R or 2S, 3R or 3S)-3-incthoxybutan-2-y1)-2-methyl- I H-pyrrolo[2,3-b]pyridine-3-earboxamide (Major Isomer Pair; Peak 2): 11-1 NMR (400 MHz, CDCI3): 8 8.179-8.163 (in, 1H), 8.143-8.120 (m, 1H), 7.558-7.531 (in, 1H), 6.986-6.954 (m, IH), 5.931 (s, 1H), 4.702-4.605 (m, 2H), 3.897 (s, 3H), 2.892 (s, 3H), 2.789 (s, 3H), 2.189 (s, 3H), 1.647-1.629 (d, 3H), 1.267-1.252 (d, 3H).
[004611 Compound 267, N44-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-((2R or 2S, 3R or 3S)-3-methoxybutan-2-y1)-2-methy1-1H-pyrrolo[2,3-b]pyridine-3-carboxamide (Minor Isomer Pair; Peak 1): 11-1 NMR (400 MHz, CDC13): 8 8.174-8.162 (d, 1H), 8.111-8.094 (d, 1H), 7.551-7.526 (rn, 1H), 6.993-6.961 (m, 1H), 5.935 (s, 1F1), 4.683-4.579 (in, 2H), 3.887 (s, 3H), 3.442 (s, 3H), 2.753 (s, 3H), 2.194 (s, 3H), 1.695-1.678 (d, 3H), 0.781-0.768 (d, 3H).
[004621 Compound 302, N-((4-methoxy-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-((2R or 2S, 3R or 3S)-3-methoxybutan-2-y1)-2-methy1-1H-pyrrolo[2,34]pyridine-3-earboxamide (Minor Isomer Pair; Peak 2): 11-1 NMR (400 MHz, CDC13): 8 8.177-8.166 (d, 1H), 8.122-8.104 (d, 1H), 7.587-7.562 (m, 1F1), 6.984-6.952 (m, 1H), 5.933 (s, 1H), 4.698-4.591 (m, 2H), 4.426 (s, 24), 3.983 (s, 3H), 3.448 (s, 3H), 2.764 (s, 31-I), 2.180 (s, 3H), 1.701-1.684 (d, 3H), 0.786-0.772 (d, 31I).
1004631 Example 66. Sµ nthesis of 14 1-ev elo n ropy I- I -methoxvpronan-2-v1)-N4(4-methoxy-6-methvi-2-oxo-1,2-dihydropyridin-3-v1)methvi)-2-methvi-I H-nvi-rolo[
2,3-.112jpvridine-3-carboxamide (Compound 283.) and its Individual Diastereonlers (Compounds 285, 286 and 292).
1004641 Step 1: N-methoxy-N-methyleyelopropaneearboxamide:
C I -.
rs;11-101 -11110.

N,0-dimethylhydroxylamine hydrochloride(i 1.2g. 114.79mmol) and TEA. (19.36g, 191.32mmol.) were dissolved in CH2Cl2 (60m1). Cyclopropanecarbonyl. chloride (10g, 95.66mino1)was added dropwise at 0 C. After addition, the mixture was allowed to warm to 25 C and stirred for! 8 h. After the reaction completed, the solution was washed with H20, saturated NaHCO3, 1 N HCI and brine. The organic layer was dried over Na2SO4 and evaporated under reduced pressure to get N-methoxy-N-methylcyclopropanecarboxamide as a yellow oil.
(8.6 g, yield 70 %).
[004651 Step 2: 1-cyclopropylpropan-1-one:
0>-<1 To N-methoxy-N-methylcyclopropanecarboxamide (2.0 g, 15.49 mmol) in THF (20 mL) was added C2H5MgBr (1.2 M in THF, 15.4 ml, 18.58 mmol) dropwise below -70 C. After addition, the solution was allowed to warm to 25 C and stirred for 18 h. After the reaction completed, the solution was quenched by addition of lmL of saturated NRIC1. Then the mixture was diluted with water and then extracted by Et0Ac. The organic layer was separated, combined, dried over Na2SO4. and evaporated under reduced pressure to get 1-cyclopropylpropan-1 -one as a white oil.
(800 mg, yield 53 %).
1004661 Step 3: 2-bromo-1-cyclopropyipropan-1-one:
Pr p- .. ...,1,1 + CtiBr2 --fi. \ , .
ei' cii,>
To 1-cyclopropylpropan-1 -one (800 mg, 8.15rnmo1) in CHC1.3 (5 mL) and EtO.Ac (5 mL) were added CuBr2 (3.64 g, 16.30 mmor.) at 70 C. The solution was stirred at 70 C
for18 h. After the reaction completed, the solution was filtered off The filtration was evaporated under vacuum to obtain 2-bromo- 1 -cyclopropylpropan-l-one as green oil. (1.0 g, yield 69%).
[00467] Step 4: Tert-butyl 1-(1.-cyclopropy1-1-oxopropan-2-yl)-2-methyl-ln-pyrrolo[2,3-blpyridine-3-carboxylate:
-A/ .0 k0 Br 0-f * 1.....<1 4 Cs2CO3 + KI --lb. nN _..1.,...1 H ----ir-41 The solution of ert-butyl 2-methyl-111-pyrrolo[2,3-14yridine-3-carboxylate (300.00 mg, 1.29 mmol), 2-bromo-l-cyclopropylpropan-l-one (342.98 mg, 1.94 mmol), Cs2CO3(841.63 mg, 2.58 mmol), K1 (42.88mg. 0.26 mmol) in .DMF(4m1) were stirred at 25 C for 18h..
After the reaction completed, the solution was partitioned between Et0Ac and H20. The organic layer was separated, dried over Na2SO4 and evaporated under reduced pressure. The residue was purified by flash column (Eluent: PE: Et0Ac=10:1) to get tert-butyl I -(1-cyclopropy1-1-oxopropan-2-y1)-2-m.ethyl-III-pyrrolo[2,3-blpyridine-3-carboxylate as a white oil. (230 mg, yield 54 %).
(00468) Step 5: Tert-butyl1-(1.-eyelopropyl-l-hydroxypropan-2-y1)-2-methyl-1.11.-pyrrolo 12, 3-bi pyridine-3-carboxylate:

o k o-/ + NaHH4 / I
N N

To tert-butyl 1-(1-cyclopropy1-1-oxopropan-2-y1)-2-methy1-1H-pyrrolo [2,3-b) pyridine-3-carboxylate (170 mg, 0.52 mmol) in CH3OH (3 ml) were added NaBHa (58.75 mg, 1.55 mmol) at 0 C. The mixture was allowed to warm 25 C with stirred for 2h. After the reaction mixture completed, the solution was concentrated under vacuum and the solid was partition between Et0Ac and H20. The organic was separated, dried over Na2SO4 and evaporated under reduced pressure to give tert-butyl 1-(1-cyclopropy1-1 -hydroxypropan-2-y1)-2-methyl-1H-pyrrolo( 2,3-b)pyridine-3-carboxyla as a white oil. (140 mg, yield 84 %).
1004691 Step 6: Tert-butyl 1-(1-cyclopropy1-1-metboxypropan-2-y1)-2-methyl-1H-pyrrolo 12, 3-bi pyridine-3-carboxylate:

NaH ¨1 Drkl HO
6\
To NaH (72.63mg, 1.82rnmo1) in THF (2mL) was added tert-butyl 1-(1-cyclopropyl-l-hydroxypropan-2-y1)-2-methyl-1H-pyrrolo[2,3-bipyridine-3-carboxyla (100 mg, 0.30 mmol) at 25 C for 30 min. To the reaction mixture was added CH31 (429.57 mg, 3.03 mmol) dropwise with stirring for 3h. After the reaction completed, the mixture was quenched by addition of 10mL of saturated NH4C1, and a clear white solution was obtained which was poured into water and extracted by Et0Ac. The extraction was dried over Na2SO4 and evaporated under reduced pressure to give tert-butyl 1-(1-cyclopropy1-1-methoxypropan-2-y1)-2-methyl-IH-pyrrolo[2,3-b]pyridine-3-carboxylate as white oil. (100mg, yield 95%).
1004701 Step 7: 1-(1-cyclopropy1-1-metboxypropan-2-y1)-2-methyl-1H-pyrrolo12,3-blpyridine-3-carboxylic acid:

o ¨et 1 / '1 e A
o 0 To a solution of tert- butyl 1-(1-cyclopropy1-1-methoxypropan-2-y0-2-methyl -1 H-pyrrolo [2,3-b]pyridine-3-carboxylate (100 mg, 0.29 mmol) in CH2C12 (2 mL) were added CF3CO2H (2 mL).The solution was stirred at 25 C for 2h. After the reaction completed, the solution was concentrated under reduced pressure to obtain 1-(1-cyclopropy1-1-methoxypropan-2-y1)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid as red oil. (87 mg, yield 99 %).
[004711 Step 8: 1-(1-cyclopropy1-1-methoxypropan-2-y1)-N-04-methoxy-6-metliy1-2-oxo-I ,2-dihyd ropyridin-3-Amethyl)-2-methyll- I.H-pyrrolo[2,3-b1pyrid ne-3-ca rb 1, a mi de (Compound 283):
.o , =
, ..2 " N I
............. N.-- 4 4 tj 4 Os, .!4.-.341;
fZi 6.
To a solution of 1-(1-cyclopropy1-1-methoxypropan-2-y1)-2-methyl-1H-pyrrolo [2, 3-b]
pyridine-3-carboxylic acid (100 mg, 0.35 mmol) in dichloromcthanc (2 mL) was added HATU
(192 mg, 0.52 mmol), TEA (105.28mg, 1.04 mmol). After stirred for 50 min at room temperature, 3-(aminomethyl)-4-methoxy-6-methylpyridin-2(1H)-one hydrochloride ( 87.5 mg, 0.52 mmol) was added. The mixture was stirred at room temperature for 3 hours.
After the reaction completed, the solution was partitioned between Et0Ac and H20. The organic was separated, dried over Na2SO4 and evaporated under vacuum.
[004721 Step 9: Separation of Isomers of 1-(1-cyclopropy1-1-methoxypropan-2-y1)-N-((4-methoxy-6-methy1-2-oxo4 ,2-d hydropy rid in-3-11)methyl)-2-me thy1-1H-pyrrolo [2,3-blpyridine-3-carboxamide (Compounds 285, 286 and 292):

1-- t>"v",-µ=sc) N SFC

HN I N...iXt \ \
0 Method 94 Compound 28$ Compound 286 Major isomer Peak 1 Peak 2 ts.
1.2 -=`;` Nr¨ SEC
H I =
41. 11 'Y = V

Method 94 Compound 292 Minor isuivier Peak "I
The residue from the previous step was solidified by MTBE to give major isomer 1 (Compounds 285 and 286) (4 mg, purity 90 %).The filtrate was purified by preparative-HPLC
(Instrument:
Gilson 281:Column: Grace C18 5u 150*25m.m ; Mobile phase A: Water (0.0225%
HCOOH
v/v); Mobile phase B: Acetonitrile (neutral); Gradient:30-60(B%);
Flowrate:22m1/min) to give major isomer 1 (285 and 286) (.30 mg, purity 100%) and minor isomer 2 (Compound 292) (2.3 mg 100%). Isomer 1 was separated by SFC (Column: AD (250*30mm, 5 um); Flow rate:
60mUrnin: Mobile: A., phase: 30% IPA+NI33.1320, B, 70 % CO2; Wavelength: 220 nm.) to give Compound 285 (15.1 mg, purity 94 %) and Compound 286 (14.5 mg, purity 97%).
[004731 Compound 285, 14(1R or S. 2R or S)-1-cyclopropyl-l-methoxypropan-2-y1)-N4(4-meth oxy-6-m ethy1-2-oxo-1,2-di hydropyri din-3-yl)methy I)-2-methyl -1H-pyrrolo [2,3-b]pyridine-3-carboxamide: LCMS (M H-1-) miz: calcd 438.23; found 439.1. 1H NMR. (400 MHz, CDC13):
6 8.112-8.162 (m, 2H.), 7.550 (t, 111), 6.933-6.964 (in, 1H), 5.931 (s, 1H), 4.609-4.708 (m, 2H), 3.761(s, 3H), 3.755-3.728 (m, 111), 3.482(s, 1H), 2.879(s,1H), 2.798 (s, 313), 2.184 (s, 3H), 1.868-1.835 (m, 1H), 1.868-1.835 (m,1H),1.764 (s,3H), 0.837(m, I.H), 0.603-0.585(d, 2H, .11...7.2Hz), 0.482-0.473(d, 211, 1004741 Compound 286, 1-((1R or S. 2R or S)-1-cyclopropy1-1-methoxypropan-2-y1)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1H-pyrrolo[2,3-b]pyridine-3-carboxamide: LCMS (M + El+) m/z: caled 438.23; found 439.2. 1H NMR (400 MHz, CDC13):
68.123-8.175 (m, 2H), 7.500 (t, 111), 6.976-7.008 (m, 1H), 5.939 (s, 1H), 4.627-4.657 (t, 2H), 4.284-4.459 (m, 1H), 3.906(s,3H), 3.795-3.814 (m, 1H), 2.86(s, 3H), 2.801 (s, 3H),2.236 (s, 3H), 1.754-1.770 (d, 3H,J=6.4), 0.844 (m, 1H), 0.589-0.606 (d.2H, j=6.8), 0.487 (m,2H).
[004751 Compound 292, 1-((1R or S. 2R or S)-1-cyclopropy1-1-methoxypropan-2-y1)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihyd ropyri di n-3-yl)methyl)-2-methyl-1H-pyrrolo [2,3-b]pyridi ne-3-carboxamide: LCMS (M + H+) mlz: calcd 438.23; found 439.2. 1H NMR (400 MHz, CDC13):
6 8.124-8.175 (m, 2H), 7.500 (t, 1H), 6.976-7.008 (m, 1H), 5.939 (s, 1H), 5.625-4.655 (t, 2H), 4.355-4.490 (m, 1H), 3.907 (s, 3H), 3.787-3.813 (m, 110, 2.879 (s, 3H),2.801 (s,3H) 2.232 (s, 3H), 1.754-1.771 (d, 3H, J=6.8),0.840-0.866 (m,2H) 0.589-0.608 (d, 2H, J=7.6), 0.487 (m,2H).
[004761 Example 67. Synthesis of ( )-N-((416-dimethyl-2-oxo-112-dihydropyridin-yOmethyl)-2-methyl-1-(1-ohenylethyl)-1H-pyrrolo12.3-ejpyridine-3-carboxamide (Compound 203) (00477) Step 1: 1-(3-methoxyphenyl)ethanol:
5ec-BuLi,EA

To a stirred solution of 3-Amino-4-picoline (7g, 64.8 mmol) in anhydrous THE
(200 mL), sec-BuLi (150 mL, 1.3M in cyclohexane, 194 mmol) was added dropwise over 20 minutes at -78 C.
The solution was warmed to room temperature and stirred at 3hours. Ethyl acetate(2.3 g, 25.9 mmol) was added dropwise into the reaction at -78 C and the mixture was stirred at the same temperature for 2 hours. Methanol (50 mL) was added dropwise into the reaction over 10 minutes. The mixture was warmed to room temperature and stirred for I hour. A
half-saturated NH4CI (250 mL) was added. The mixture was extracted with EA. The combined organic layers were washed with brine, dried and concentrated to afford the crude product.
The crude product was purified by silica gel chromatography (petroleum ether / ethyl acetate =
10:1) to afford 2-methy1-1H-pyrrolo[2,3-c]pyridine (2.5 g, 73.5%).
1004781 Step 2: 2,2,2-trichloro-1-(2-methyl-1H-pyrrolo(2,3-clpyridin-3-yl)ethanone:
CI CI NH
AIC13.C13C0CI

To a stirred solution of 2-methy1-1H-pyrrolo[2,3-c]pyridine (2.5 g, 18.9 mmol) and aluminum chloride (5 g, 37.8 mmol) in DCM (100 mL), trichloroacetylchloride (4.1 g, 22.7 mmol) was added dropwise into the reaction over 0.5 hours at room temperature. After stirring 2 hours, the reaction was cooled to 0 C and was quenched with water (100 mL). The resulting precipitate was isolated by filtration to afford 2,2,2-trichloro-1-(2-methy1-111-pyrrolo[2,3-c]pyridin-3-yl)ethanone which was used for next step without further purification. Assumed 100% yield.
(5.24g).
1004791 Step 3: Methyl 2-methyl-1H-pyrrolo12,3-c I py ridine-3-carboxylate:
Ci NH
CI CI NH KOH
.......................
o L.,N
A mixture of 2,2,2-trichloro-1-(2-methy1-1H-pyrrolo[2,3-c]pyridin-3-ypethanone (5.24 g, 18.9 mmol) and KOH (1.2 g, 20.9 mmol) in Me0H (100 mL) was stirred at room temperature for 16 hour. The reaction mixture was concentrated to remove Me011, the residue was partitioned between EA and Water. The organic layer was washed with brine, dried and concentrated to afford methyl 2-methyl-1H-pyrrolo[2,3-c]pyridine-3-carboxylate (3 g, 83%).
1004801 Step 4: Methyl methyl 2-methyl-1-(1-phenylethyl)-1H-pyr ro I o [2,3-cip ri di It e-3-earboxylate:
_______________________ 0 /
/

A mixture of methyl 2-methyl-1H-pyrrolo[2,3-c]pyridine-3-carboxylate (550 mg, 2.89 mmol) and sodium hydride (200 mg, 4.34 mmol) in N,N-dimethylformamide (3.0 inL) was stirred at room temperature for 0.5 hour, and then (1-bromoethypbenzene (589 mg, 3.18 rnmol) was added. The mixture was stirred at room temperature for 3 hours. The reaction mixture was poured into saturated NII4C1 and extracted with ethyl acetate. Organic layers were combined and concentrated to give a residue. The residue was purified by chromatography (petroleum ether /
ethyl acetate = 5:1) to give methyl 2-methy1-1-(1-phenylethyl)-1H-pyrrolo[2,3-c]pyridine-3-carboxylate (800 mg, 94%).
1004811 Step 5: 2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-clpyridine-3-carboxylic acid:
\k"
HO
w WA.

CM
f4 14 N 0 7.

To a mixture of methyl 2-methyl-I -(1-phenylethyl)-1H-pyrrolo[2,3-e]pyridine-3-carboxylate (800 mg, 2.72 mmol) and [(OH (1.5 g, 27.2 mmol) in (15 mL) and water (5 mL) was refiuxed for 2 hours. The mixture was adjust PH to 2 by 10% HC1 and extracted with EA. The combined organic layers were washed with brine, dried and concentrated to afford the crude product. The crude product was used into the next step without more purification. 100%
yield. (760 mg).
[00482] Step 6: ( )-N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-e] py rid in e-3-carboxamide (Corn pound 203):
\
µ)"..0 I H I
A mixture of 2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-c]pyridine-3-carboxylic acid (280 mg, 1.0 mmol) was added HATU (456 mg, 1.2 mmol), TEA. (1 g, 10 mmol) and 3-(aminomethyl)-4,6-dimethylpyridin-2(I H)-one (182 mg, 1.2 mmol) in anhydrous dichloromethane (30 mL) was stirred at room temperature for 16 hours. To the reaction mixture was added water (10 ML), extracted with dichloromethane (30 mL x 2). The organic layers were combined and concentrated to give a residue. The residue was rereystal.lized from MeCN to affotd compound N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-c]pyridine-3-carboxamide as an off-white solid (80 mg, 21.6%).
LRMS (M+11 ) calcd 414.21; found 414. 1H NMR (400 MHz, Methanol-d4) 6: 8.84 (s, 111), 8.16 (d, J= 7.6 Hz, 1H), 8.03 (d, J= 6.8 Hz, 1H), 7.44-7.37 (m, 5H), 6.09 (s, 111), 6.01-5.99 (m, 1H), 4.49 (s, 2H), 2.73 (s, 3H), 2.38 (s, 3H), 2.22 (s, 3H), 2.06 (d, J= 7.2 Hz, 31-1).
[00483] The compounds shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
Structures are shown in Figure 1.
Compound Name NMR mlz ( )-N-((4-ethoxy-6- NMR (400 MHz, CDC13) 6: 8.34 (s, 1H), 8.11-methy1-2-ox.o-1 ,2- 8.34 (d, J= 5.6 Hz, 1H), 7.75-7.82 (m, 2H), 255 d ihydropyridi n-3- 7.26-7.32 (m, 211), 7.13-7.16 (d., J= 7.2 Hz, yOmethyl)-2-methyl-1.- 2H), 5.85-5.90 (m, 2H), 4.68-4.70 (m, 2H), 445 (1. -phenylethyl)-1H- 4.10-4.16 (m, 211), 2.80 (s, 311), 2.14(s, 3H), pyrrolo[2,3-e]pyridine-3- 1.96-1.99 (dõ I... 7.2 Hz, 311), 1.44-1.48 (m, carboxamide 3H).

( )-N-((4 6-dimethy2 (400 MHz, CHLOROFORM-d) 8 ppm 1.63 (br.
,1--s., 3 F1) 2.21 (s, 3 Fl) 2.41 (s, 3 El) 2.73 (s, 3H) oxo-12-dihydropyridin-3.24 (s, 3 H) 3.72 (dd, J=9.81, 5.40 Hz, 1 H) 3-yOmethyl)-1-(1-240 3.80 - 3.88 (m, 1 H) 4.60 (d, J=5.95 Hz, 2 H) methoxypropan-2-v1)-2-4.71 (dd, J=13.23, 7.06 Hz, 1 H) 5.92 (s, 1 El) 383 methy1-1H-pyrrolo[2,3-7.31 (d, J=5.73 Hz, 1 H) 7.38 (br. s., 1 H) 8.26 elpyridine-3-(d, J=5 .29 Hz, 1 H) 9.09 (br. s., 1 H) 11.07 (br.
carboxamide s., 1 H) ( )-N-((4-methoxy-6- (400 MHz, CHLOROFORM-d) 8 ppm 1.62 (br.
methyl-2-oxo-1,2- s., 3 H) 2.26 (s, 3 H) 2.75 (s, 3 H) 3.25 (s, 3 H) dihydropyridin-3- 3.72 (dd, J=9.81, 5.40 Hz, 1. H) 3.80 -3.87 (m, yOmethyl)-2-methyl-1- I H) 3.90 (s, 3 H) 4.65 (d, J=5.29 Hz, 2 H) 4.71 431 (1-phenylethyl)-1H- (dd, J=13.78, 6.95 Hz, 1 H) 5.93 (s, I H) 7.32 pyrrolo[2,3-c]pyridine-3- (br. s., 1 H) 7.50 (br. s., 1 H) 8.25 (br. s., 1 H) carboxamide 9.11 (br. s., I H) [00484] Step 7: Chiral separation of N-((4,6-dimethy1-2-oxo-1,2-dihydropyridin-yl)methyl)-2-methy1-1-(1-phenylethyl)-1H-pyrrolo [2,3-c] pyridine-3-ca rbox amide isomers (Compounds 247 and 248):
oN
cl\N SFC
3 Compound 247 Peak liNt Compound 248 Peak 2 [00485] Compound 203, 45 mg was separated by SFC (Column: Chiralpak AD
(250*30mm,5um); Flow rate: 50mL/min: Mobile: A, phase: 35% IPA+NH3.H20, B, 65 % CO2;
Wavelength: 220 nm) to give (R or S)-N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl- I -(1-phenylethy1)-1H-pyrrolo[2,3-c]pyridine-3-carboxamide (Peak 1;
Compound 247;
12 mg, purity 98 A) and (R or S)-N4(4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-141 -phenylethyl)-1H-pyrrolo [2,3-c]pyridine-3-carboxamide (Peak 2;Compound 248; 11 mg, purity 98%).
[00486] Compound 247, (R or S)-N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl.-1-(1-phenylethyl)-1H-pyrrolo[2,3-c]pyridine-3-carboxamide: LRMS (M
H4.) m/z: calcd 414.21; found 414. IH NMR (400 MHz, CD30D): 8 8.61 (s, 111), 8.25-8.2.1 (q, J=6.8 Hz, 211), 7.41-7.36 (m, 3H), 7.35-7.27 (m, 211), 6.60 (s, 1H), 6.28-6.23 (q,1=6.8 Hz, 1H), 4.61 (s, 2H), 2.81 (s, 3H), 2.53 (s, 3H), 2.38 (s, 3H), 2.05-2.03 (dõ/=7.2 Hz, 3H).
[004871 Compound 248, (R or S)-N44,6-dimethyl-2-oxo-1,2-dihydropyridin-3-y1)methyl)-2-methyl-1-(1-phenylethyl)-1H-pyrrolo[2,3-c]pyridine-3-carboxamide: LRMS (M H4) m/z: calcd 414.21; found 414. 1H NMR (400 MHz, CD30D): 6 8.63 (bs, 1H), 8.27-8.22 (q, J=6.4 Hz, 211), 7.42-7.37 (m, 3H), 7.36-7.28 (m, 2H), 6.80 (s, 1H), 6.29-6.24 (q, J=6.8 Hz, 111), 4.65 (s, 2H), 2.82 (s, 3H), 2.59 (s, 3H), 2.45 (s, 3H), 2.06-2.04 (d, J=7.2 Hz, 3H).
(00488) The compounds shown in the following table were prepared according to the general procedure outlined in this example using the appropriate starting materials and modifications.
Structures are shown in Figure 1.
Compound Name NMR raiz (4(X) MHz, CDCI3) 6 12.2-12.1 (br, 1H), (R or S)-N-((4-methoxy-6-8.29-8.27 (d, J= 5.2 Hz, 1H), 8.07-8.06 methy1-2-oxo-1,2-(d, J= 5.2 Hz, 1H), 7.70-7.62 (in, 2H), 258 2 dihydropyridin-3-yl)methyl)-7.26-7 21 (m, 3H), 7.08-7.06 (dõ/ = 431 - -phenyet )-Hz /111 ) 5.86 (s, 11-1), 5.82-5.76 (q, -methy1141 l hyl .
1 H-pyrrolo[2,3-clpyridine-3-7./ Hz, 1H,), 4.61-4.60 (d, j= 4.0 Hz, 2H), carboxamide 3.83 (s, 3H), 2.73 (s, 3H), 2.14 (s, 3H), 1.91-1.89 (d, J = 7.2 Hz, 3H) (400 MHz, CDCI3) 6 8.28-8.27 (d, J = 5.2 (R or S)-N-((4-methoxy-6- Hz, 1H), 8.07-8.06 (d, J= 5.2 Hz, 1H), methyl-2-oxo-1,2- 7.70-7.62 (m, 2H), 7.26-7.21 (m, 3H), dihydropyridin-3-yl)methyl)- 7.08-7.06 (d, J= 7.2 Hz, 2H), 5.86 (s, 2-methyl-1-(1-phenylethyl)- 1H). 5.82-5.76 (q, J = 7.2 Hz, 1H),4.61-1H-pyrrolo[2,3-c]pyridine-3- 4.60 (dd, Jj = 1.6 Hz .12 = 4.0 Hz, 2H), carboxamide 3.83 (s, 3H), 2.73 (s, 3H), 2.14-2.13 (d, J
PEAK 2 4.0 Hz, 3H), 1.91-1.89 (d, J= 6.8 Hz, 3H) (00489) Example 68. Synthesis of 3-(aminomethyl)-4-methoxy-6-methylpyridin-2-ol: The title intermediate was synthesized according to the following Scheme:

1) NaH, THF, 0 C, 1 h CN 10% HCI, reflux, 4 h C N
NC.CN __________ 11 I I
2) fl -10 c. 1 h 0 NH, N OH
Cl PC1C12. reflux CN MeONa, methanol, 3 h (r' reflux, 4 h II
N CI

=-=..
H2. Boe20, TEA, Raney Ni. I 4 N HCI in H20 I
methanol. THF, r 11 .t., overnight " reflux. 4.5 h r'r'NH2 [004901 Step 1: 2-amino-6-methyl-4-oxo-4H-pyran-3-carbonitrile: Malononitrile (110 g, 1.67 mol) was dissolved in dry THF (1000 ml) and cooled in ice-water bath.
Na.H (60 % in mineral oil, 67 g, 1.67 mol) was added portionwise below 10 C very carefully while the reaction flash was evacuted with N2 flow. After addition completed, the mixture was stirred at 0 C for 30 min. Then 4-methyleneoxetan-2-one (140 g, 1.67 mol) was added dropwise below 0 C. After addition completed, the mixture was stirred at -10 C for I h. The reaction mixture was neutralized by 4 N MCI and concentrated under vacuum to give compound 2-amino-6-methy1-4-oxo-4H-pyran-3-carbonitrile as an orange oil. The crude product was used to next step without further purification.
1004911 Step 2: 2,4-dihydroxy-6-methylnicotinonitrile: 2-amino-6-methy1-4-oxo-pyran-3-carbonitrile from above was dissolved in 4 N HCl/H20 (2500 ml) and refluxed for 5 h with stirring strongly. After cooled to r.t., the precipitate was filtered, washed with H20 (500 ml), ethanol (500 ml) and MTBE (200 ml) and dried under high vacuum. 2,4-dihydroxy-6-methylnicotinonitrile was obtained as a yellow powder. (165 g, yield 66 %).
[004921 Step 3; 2,4-dichloro-6-methylnicotinonitrile: 2,4-dihydroxy-6-methylnicotinonitrile (40 g, 266.4 mmol) was dissolved in POCI3 (120 ml) and added by DMF (4 drops).
The mixture was heated for 3 h. Then the mixture was concentrated under vacuum. The residue was dissolved in Et0A.c (2 L) and neutralized by saturated NaHCO3. Then the mixture was filtered through a Celite pad to remove the dark flocculating. The organic layer was separated, dried over Na2SO4 and concentrated under vacuum to give 2,4-dichloro-6-methylnicotinonitrile as an off-white solid. (45 g, yield 90 %).

[004931 Step 4: 2,4-dimethoxy-6-methylnicotinonitrile: 2õ4-dichloro-6-methylnicotinonitrile (45 g, 240 mmol) was dissolved in CH3OH (300 ml). Na0Me (30 % in Me0H, 100 ml, 1680 mmol) was added. The mixture was refluxed for 4 h. After cooled to r.t., the reaction mixture was neutralized by HOAc. The solvent was removed under vacuum and the residue was washed with H20 (300 ml) and MTBE (100 ml). The resulting solid was coevaporated with dry THF (300 ml) to give 2,4-dimethoxy-6-methylnicotinonitrile as a dark-yellow solid. (40 g, yield 95 %).
1004941 Step 5: tert-butyl ((2,4-dimethoxy-6-methylpyridin-3-Amethyl)carbamate: 2,4-dimethoxy-6-methylnicotinonitrile (10.0 g, 56 mmol) was dissolved in the mixture of THE (260 ml) and methanol (260 ml). Raney Ni (wet, 10.0 g), TEA (29.0 g, 280 mmol) and Boc20 (36.8 g, 168 mmol) were added. Then the mixture was hydrogenated (1 atom.) at r.t.
overnight. After reaction completed, the reaction mixture was filtered through a Celite pad. 6 parallel reactions were combined and concentrated under vacuum to give tert-butyl ((2,4-dimethoxy-methylpyridin-3-yl)methyl)carbamate as a yellow solid. (84 g, yield 88 %).
[004951 Step 6: 3-(aminomethy1)-4-methoxy-6-methy1pyridin-2-o1: tert-butyl ((2,4-dimetboxy-6-methylpyridin-3-yOmethyl)carbarnate (83 g, 294 mmol) was dissolved in 4 N
FICl/H20 (830 ml). Then the mixture was refluxed for 4.5 h. (The reaction mixture was monitored by MS spectrum to make sure the methyl group at 2-position de-protect completely.) After the reaction completed, the mixture was concentrated under vacuum to give a brown oil.
The oil was suspended in Etal (300 ml) for 15 min to give a yellow precipitate. The precipitate was filtered, washed with ethanol (100 ml) and MTBE (100 ml) and dried under high vacuum to give 38 g of fraction 1 3-(aminomethyI)-4-methoxy-6-methylpyridin-2-ol (Purity 98 % by LCMS, yield 63 %) as a yellow powder. In the meantime, the filtration from fraction 1 was concentrated under vacuum and the residue was solidified by ethanol (100 ml).
The precipitate was filtered, washed with ethanol (100 ml) and MTBE (100 ml) and dried under high vacuum to give 20 g of fraction 2 3-(aminomethyl)-4-methoxy-6-methylpyridin-2-ol (Purity 94 % by LCMS, yield 33 %) as a yellow powder.
1004961 Example 69. Synthek4 of 3-(a ntinometin 1)-4-etird-O-Inet h ritlin-2t I ft)-one.
The title intermediate was synthesized according to the following Scheme:

0 NC, )1.
1)PPh3,0E-101.1 11)11 ph o CN

1) tBuOK
P:Ph --------- ------ *.^ I
C I 2)3M NaOH 48 h -B c 2H2 Ni,H2(i atm) CC Ha/dioxane I HCI
Eloc.20,NEt3 N 0 N f 0 [004971 Step 1: 1-(triphenylphosphoranylidene)propan-2-one: A solution of 1-chloropropan-2-one (50 g, 540.4 mmol) in chloroform (150 mL) was added dropwise to a solution of triphenylphosphine (141.72 g, 540.4 mmol) in chloroform (150 mL) under nitrogen.
The mixture was stirred at 70 C for 12hr, and the resulting phosphonium salt was filtered. The precipitate was washed with ethyl acetate and dried under vacuum. The dried phosphonium salt was suspended in a mixture of water (250 mL) and methanol (250 mL), and the mixture was stirred for 1 hr. Aqueous sodium hydroxide (2.00 M) was added to the mixture until a pH
between 7 and 8 was reached. The mixture was then stirred vigorously for 1 hr.
The phosphorane precipitate was filtered and washed with water. After drying in vacuum., the phosphorane was recrystallized from ethyl acetate and dried under vacuum to afford 1-(triphenylphosphoranylidene)propan-2-one (40.00 g, 23.3%) as a white solid.
[004981 Step 2: hex-3-en-2-one: To a solution of 1-(triphenylphosphoranylidene)propan-2-one (40 g, 125.65 mmol) in dichloromethane (150 mL) was added propionaldehyde (45.83 g, 789.07 mmol) at 24 C. The reaction mixture was then stirred at 24 C for 12 hr. After concentration, the residue was then distilled under vacuum (73 C/-0.09 MPa) to give hex-3-en-2-one (5.36 g, 43.5%).
[004991 Step 3: 4-ethyl-6-methy1-2-oxo-1, 2-dilitydropyridine-3-carbonitrile:
To a stirred solution of potassium 2-methylpropan-2-olate (4.92 g, 43.81 mmol) and 2-cyanoacetamide (4.05 g, 48.19 mmol) in (methylsulfinyl)methane (60 mL) was added hex-3-en-2-one (4.30 g, 43.81 mmol) under nitrogen atmosphere at 25 C. The reaction mixture was then stirred at 25 C for 30 min, and then additional potassium 2-methylpropan-2-olate (14.75g, 131.44 mmol) was added.
Nitrogen gas was displaced by oxygen gas and the mixture was stirred at 25 C
for 48 hr. The mixture was diluted with 4 volumes water (240 mL), and then 5 volumes of 4 N
HCI (300 mL), which were added slowly. The reaction mixture was filtered, washed with water, and dried to give 4-ethyl-6-methyl-2-oxo-1, 2-dihydropyridine-3-carbonitrile (1.30g. 18.3%) as a gray solid.
[005001 Step 4: tert-butyl ((4-ethyl-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)carbamate: To a solution of Raney Ni (0.8 g) in methanol /
tetrahydrofuran (72 mL, 1 / 1) was added 4-ethyl-6-methyl-2-oxo-1,2-dihydropyridine-3-carbonitrile (1.20 g, 7.40 mmol) , triethylamine (1.50 g, 14.80 mmol) and di-tert-butyl dicarbonate (1.94 g, 8.88 mmol). The reaction mixture was stirred at 23 r under hydrogen pressure (I atm) for 20hr.
The reaction mixture was filtered through Celite. The filtrate was diluted with ethyl acetate, washed with brine and dried over anhydrous sodium sulfate. The solvent was then removed under vacuum to afford crude tert-butyl ((4-ethyl-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)carbamate ( 1.46 g, 71.2%) as a white solide for the next step.
[005011 Step 5: 3-(aminomethyl)-4-ethyl-6-methylpyridin-2(1 H)-one: Tert-butyl ((4-ethyl-6-methy1-2-oxo-1,2-dihydropyridin-3-yl)methyl)carbamate (1.00 g, 3.75 mmol) was dissolved in a solution ofhydrogen chloride in1,4-dioxane (20 mL). The mixture was stirred for 2hr. The reaction mixture was filtered. The residue was washed with dichloromethane, dried to afford 3-(aminomethyl)-4-ethy1-6-methylpyridin-2(1H)-one hydrochloride (593 mg, 77.9%) as a light yellow solid. LRMS (M + 1-1-) m/z: calcd 166.11; found 167.1. 111 NMR (400 MHz, D70): 5 ppm 6.31 (s, I H), 4.06 (s, 2 1-1), 2.57 (q, J=7.86 Hz, 2 H), 2.25 (s, 3 H), 1.10 (t, J = 7.53 Hz, 3 H).
[005021 Example 70. Synthesis of 34aminome1hy1)-44difinoromethoxv)-6-methylps ridin-2(11-1)-one. The title intermediate was synthesized according to the following Scheme:

N. ....... P003, reflux 1. AN.... .0 Cs0Ac,DIVIF . ,....
CN meoNameoli Lx.,CN
N.. 0H
...,..exCH
,..1N-S.CCI ii "....CN-... CI i)Its r..."..õ.
.-= 0...-N
OCT _HF2 2 ocHF, 0 , _ 0 , _ HC
F..1."F
47,..,o.:CN __________________________ 0K...
H2,Raney Ni H HIr : NH2 NaH2ONIF N 0. N 0 [005031 Step 1: 2,4-dichloro-6-methylnicotinonitrile: To a solution of 2,4-dihydroxy-6-methylnicotinonitrile (20.0g, 133.0mmo1) in POC13 (150mL) was stirred at 120 C for 2 hours under N2. It was partitioned between water (500 mL) and ethyl acetate (500 mL), the organic layer was dried and concentrated to afford 2,4-dichloro-6-methylnicotinonitrile (15.3g brown soild, 61.4% yiled).
[005041 Step 2: 2-ehloro-4-hydroxy-6-methylnicotinonitrile: A mixture of 2,4-dichloro-6-methylnicotinonitrile (12.0g, 64.2 mmol ) , C:s0Ac ( 37.0g, 193.0 mmol) in N,N-dimethylformamide (50 mL) was stirred at 80 C overnight under N2. The mixture was partitioned between water(800 mL) and ethyl acetate (800 mL), the organic layer was dried and concentrated to afford 2-chloro-4-hydroxy-6-methylnicotinonitrile (9.0g brown solid, 84.1 %
yiled).
[005051 Step 3: 4-hydroxy-2-methoxy-6-methylnicotinonitrile: A mixture of 2-chloro-4-hydroxy-6-methylnicotinonitrile (2.0g, 11.9 mmol) , sodium methanolate ( 3.2g, 59.5 mmol) in methanol (20 mL) was stirred at 60 C overnight under N2. The mixture was quenched with HC1 (1M) to pH=2. It was partitioned between water(500 mL) and ethyl acetate (500 mL), the organic layer was dried and concentrated to afford 4-hydroxy-2-methoxy-6-methylnicotinonitrile (2.0g brown solid, 100 % yiled).
[005061 Step 4: 4-(difluorometboxy)-2-methoxy-6-methylnicotinonitrile: To a solution of 4-hydroxy-2-methoxy-6-methylnicotinonitrile (2.0g, 12.2 mmol) in N,N-dimethylformamide(10 mL) was added sodium hydride (880 mg, 36.6 mmol) at 0 'C and the mixture was stirred for 0.5 hour, Ethyl 2-chloro-2,2-difluoroacetate ( 5.4g, 39.0 mmoI) was added with vigorous stirring, over the course of 20 mm. The suspension was warmed to 80 'C overnight under N2. The mixture was quenched into Na2CO3 (200 mL). It was partitioned between water(500 mL) and ethyl acetate (500 mL), the organic layer was dried and concentrated to afford the crude, which was purified by flash column (PE:EA=20:1) to afford 4-(difluoromethoxy)-2-methoxy-6-methylnicotinonitrile (550 mg yellow solid, 22.0% yiled).
[005071 Step 5: tert-b u tyl ((4-(difluoromethoxy)-2-methoxy-6-methylpyridin-3-yl)methyl)carbamate: To a solution of 4-(difluoromethoxy)-2-methoxy-6-methylnicotinonitrile (550mg, 2.58mmo1), di-tert-butyl dicarbonate (844 mg, 3.87 mmol), Triethylamine (391 mg, 3.87 mmol) and Raney Ni (2g) in Tetrahydrofuran(10mL) was stirred at room temperature overnight under H2. It was filtered and the filtrate was concentrated to afford tert-butyl((4-(difluoromethoxy)-2-methoxy-6-methylpyridin-3-yl)methyl)carbamate. (830mg yellow solid, 100 % yiled).

1005081 Step 6: 3-(aminomethyl)-4-(difluoromethoxy)-6-methylpyridin-2(1H)-one:
To a solution of tert-butyl((4-(difluoromethoxy)-2-methoxy-6-methylpyridin-3-yl)methyl)carbamate (830mg, 2.61mmol) in HC1(10mL) was stirred at 100 'C for 1.5 hours under N2.
The mixture was concentrated to afford 3-(aminomethyl)-4-(difluoromethoxy)-6-methylpyridin-2(1H)-one(430mg,yellow solid, 80.8 % yield). LCMS (M + H+) m/z: calcd 204.07; found 205Ø 111 NMR (400 MHz, DMS0): 6 7.621-7.258 (t ,1H), 6.26 (s, 111), 3.837-3.822 (d, J =
6.0Hz,2H) 1005091 Example 71. Synthesis of 3-(aminoinethyl)-44-dimethylpyridin-2(111)-one. The title intermediate was synthesized according to the following Scheme:
HCl/1.4-0 0 -"" I
CN HN
HN I NN2 -NCI Ni1112 I LI
dioxane `80c 0 Me0H 0 0 1005101 Step 1: 4,6-dimethy1-2-oxo-1,2-dihydropyridine-3-carbonitrile: To a solution of pentane-2,4-dione (100 g, 1.0 mol) in H20 (2 L) were added 2-cyanoacetamide (84 g, 1.0 mol) and K2CO3(i 3.8 g, 0.1 mol). Then the mixture was stirred at room temperature for 16 hr. The reaction solution was filtrated to give crude product. The crude was washed with water and concentrated to give 4,6-dimethy1-2-oxo-1,2-dihydropyridine-3-carbonitrile (138 g, 93 %).
1005111 Step 2: tert-butyl ((4,6-dimethy1-2-oxo-1,2-dihyd ro pyridin-3-yl)methyl)ca rba mate: To a solution of 4,6-dimethy1-2-oxo-1,2-dihydropyridine-3-carbonitrile (40 g, 0.27 mol) in THFCH3OH (1:1, 2 L) were added Ni (40 g), Boc20(110 g, 0.5 mol) and Et3N(50 g, 0.5 mol). Then the mixture was stirred in H2 atmosphere at room temperature for 48 hr. The reaction solution was filtrated and concentrated to give crude product. The crude was added H20 (200 mL) and extracted by DCA (600 mL*3). The organic layer was concentrated to give tert-butyl 04,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yOmethypcarbainate (40 g, 56 %) for next step.
1005121 Step 3: 3-(aminomethyl)-4,6-dimethylpyridin-2(11I)-one: tert-butyl ((4,6-dimethy1-2-oxo-1,2-dihydropyridin-3-yOmethypearbamate (40 g, 0.27 mol)was added into dioxanellICI (1 Wand the mixture was stirred at room temperature for 4 hr. The reaction solution was filtrated and concentrated to give crude product. The crude was washed with ethyl acetate (100mL*2) and Et0H (50mL*1) and concentrated to give 3-(aminomethyl)-4,6-dimethylpyridin-2(1H)-one hydrochloride (15 g, 40 %). LCMS (M + H+) miz: calcd. 152.19; found 153.1. 111 (DMSO, 400MHz) 8 11.84 (s, 1H), 8.07 (s, 3H), 5.96 (s, 1H). 3.76-7.75(d, J =
5.6 Hz, 2H), 2.21 (s, 3H), 2.15 (s, 3H).
[005131 Examnle 72. General Procedures for Svnthesizine Other Comnounds of the Invention 100514[ Gemend Procedure ,A.: lath& Alk'sotion y--0/1"le -We 1 ''x...... \ + Rx-Br ..-\/.
A N NaH CAW ., #....õ, c H Fix NH indoie ester RX
Alkylatad indole ester To a cooled (0 C) solution of iVH indole ester (I equivalent) in N,N-dimethylformamidc (volume to make concentration 0.4M) was added sodium hydride (60% w/w, 1.1 equivalents relative to indole). The resultant mixture was stirred for 15 minutes. Then RX (2 equivalents) was added and the reaction was allowed to warm to room temperature. The reaction was maintained at ambient temperature for 12 hours. The reaction mixture was poured into saturated ammonium chloride solution (100 mL) with stirring. The mixture was extracted with ethyl acetate (200 mL x 2) and the combined organic phase was washed with brine, dried over magnesium sulfate, filtered, and concentrated to give crude product which was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 20:1) to afford the desired alkylated Indole ester product.
100515] General Procedure B: Saponification of alkylated Indole ester o o Li0H, THF/Nie0H/H20 OH
- I \
A-- N A NI, Alkylated indole ester Indoie acid To a solution of alkylated Indole ester (I equivalent) in tetrahydrofuran:methanol:water (2.5:5:1, volume to make concentration 0.05M) was added lithium hydroxide (4 equivalents). The resultant reaction mixture was stirred at 60 C for 48 hours. The mixture was concentrated in vacuo. Then the residue was diluted with water (40 mL) and slowly acidified with IN hydrogen chloride to pH = 4-5. The mixture was extracted with ethyl acetate (100 mL x 3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated to give crude indole acid, which was used in the subsequent step without additional purification.
1005161 General Procedure C: Amide bond formation Ria EDCI, HOBT 9 Et3N, CH2Cl2 N /
\ A
I Xtr--I H
A 1St tsc OH OH
Rx Indole acid Pyridone amine Target To a solution of lndole acid (1 equivalent) in dichloromethane (volume to make concentration 0.05M) were added 1-hydroxybenz,otriazole (1.5 equivalents), 1-(3-dimethylaminopropyI)-3-ethylcarbodiimide hydrochloride (1.5 equiv.) and triethylamine (3 equiv.). The resultant mixture was stirred at room temperature for 30 minutes. Then Pyridone amine (1.2 equiv.) was added and the resultant mixture was stirred at room temperature for 16 hours. Water (50 mi.) was added to the mixture. The mixture was extracted with dichloromethane (100 mt, >, 2).
The organic layer was concentrated in vacuo to provide crude product which was purified by column chromatography (silica gel, dichloromethane/ methanol = 20:1) to afford the target compound.
1005171 General Procedure D: Chiral chromatography Separation of chiral compounds was accomplished via normal phase HPLC or SFC
(supercritical carbon dioxide fluid chromatography). Separated compounds were typically >95%
cc. The absolute configuration of chiral centers was not determined.
1005181 General Procedure E: Amination of pyrrolopyrimidines Rx Ray' Rx la R
amine r N N / Niv N N / R20 Oti 0 0 --sts1 =
R2.
Chloropyrimidine R20 and R21 are independently Aminopyrimidine any appropriate nitrogen substituent or R20 and R21 are taken together with the nitrogen atom to which they are bound to form a heterocyclic ring A solution of Chloropyrimidine (1 equiv.) in amine (volume to make concentration 0.1M) was stirred at 150 C for 30 minutes under microwave (pressure: 12.2 bar, equipment power: 150W).
The mixture was concentrated in vacuo and purified by column chromatography (silica gel, dichloromethanelmethanol = 10: 1) to afford the target aminopyrimidine.

1005 l91 General Procedure F: Suzuki coupling of chloropyrimidine --I¨ 0 . Rx Rx rt/
R2.,21,k14 N

pd(cippOc12 0 ¨NP¨R23 0 ¨N
Chloropyrimidine Coupled product H R17r R 1.( 22 = or NAPEt R23 = aryl. beteroaryl, partially saturated carbocyclyl, partially satruated beterocycly, 6H alkenyl or any moiety containing a vinyl group Chloropyrimidine (1 equiv.), boronic ester or acid (1.4 equiv.), Pd(dppf)C12(0.1 equiv.), K2CO3 (2 equiv.) were combined in 3:1 dioxane:water (volume to make concentration 0.2M), then stirred at 140 C under microwave irradiation for 30 mins. The mixture was filtered through celite, concentrated in vacuo and purified by column chromatography to afford the target coupled product.
1005201 General Procedure C: Hydrogenation of Coupled product Rx Rx õsik-4 Pd/C, WON

N
23a 0 ¨N
IR
Coupled product R23a = hydrogenated version of Pd/C (catalytic) was added to a solution of Coupled product (1 equiv) in Me0H
(volume to make concentration 0.1 M) and stirred at r.t for 4h. The mixture was concentrated in vacuo and purified by preparative-HPLC to afford the desired product.
1005211 General Procedure H: Pd-catalyzed methylation of chloropyrimidine ethyl ester Ps' \
AlMe3,Pd(PPhil) r_Nõ
0 )C5x¨N THF,reflux 0 Chloropyrimidine ethyl ester Methyl pyrimidine ethyl ester Al(CH3)3 (2 equiv.) was added dropwise under N2 at 20 C through a septum to a stirred solution of (R)-ethyl 2-chloro-6-methy1-7-(1-phenylethyl)-7H-pyrrolo [2,3-d] pyrimidine-5-carboxylate(400 mg, 1.2 mmol) in 20 ml THE and Pd(PPh3)4 (63 mg, 0.06 nunol).
The mixture was then stirred at 70-80 'C for 8 h, then cooled to RT and poured into saturated aqueous NH4CL'ice and filtered. The filtrate was washed with dichloromethane (3X), then the combined organic phases were dried (Na2SO4), filtered and concentrated. The material was purified by silica gel column chromatography.
[005221 General Procedure I: Hydrolysis of nitrile to acid LOH
Rx Rla R'a NH )X., Nti/ Ntt CHIOH HN H N 0 0 """ N
Nitrile Acid Lithium hydroxide anhydrate (10 equiv.) in water (2 mL) was added to Nitrite (1 equiv.) in methanol (10 ml.,) and the resultant mixture was stirred at room temperature for 12 hours. The mixture was evaporated, added with water (5 mL), acidified with aqueous hydrochloric acid (1M) to pH = 2. The solid precipitate was filtered and dried to obtain Acid.
[005231 General Procedure J: Methyl ester formation Wa P4 R1 a H2SO4 H -~COOH CH30H N11,11/ N
H N N
70 C ,--COOCH3 , Acid Methyl Ester To a solution of Acid (1 equiv.) in CH3OH (Volume to make concentration 0.1 M) was added with H2SO4 (2 equiv.) and the reaction mixture was stirred at 70 C for 2 hours,. The solution was concentrated, diluted with water (20 mL), extracted with ethyl acetate (20 mL). The organic layers were separated, combined, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue Ester, which was used directly in the next reaction.
[005241 General Procedure K: Reduction of methyl ester to alcohol.

fel Rx N
HN I
HN 11,....kN NaBH4 k )--COCICH3 ;NOM
0 ¨N
Methyl Ester Alcohol To a solution of Methyl ester (1 equiv.) in Et0H (volume to make concentration 0.1M) was added with NaBH4 (10 equiv.), and the reaction mixture was stirred at 80 C
for 12 hours. After the completion of the reaction, the reaction was quenched by addition of 5 mL
of saturated aqueous NH4C1 solution, and extracted with Et0Ac (3X). The combined organic washes were washed with H20, then dried (Na2SO4), filtered and concentrated. Purification by RPHPLC
provided the target compound.
[005251 General Procedure L: Sulphonylation 0 ,o HN N N +
tiN ' Chiral amine To a solution of Chiral amine (1 equiv.) in dichloromethane (volume to make concentration 0.1 M) was added triethylamine (4 equiv.) at 18 C under N2. The reaction was cooled to 0 C and methanesulfonyl chloride (1.5 equiv.) was added. The reaction was stirred at 0 C for 1 h. Then the mixture was concentrated in vacuo and methanol and potassium carbonate were added and the reaction was stirred for another 1 h. The mixture was filtered and the crude product was purified by preparative-HPLC.
[005261 The table below lists compounds of the invention and which of the above general methods was used in their synthesis. Structures of these compounds are set forth in Figure 1.
General Methods Compound Name NMR data miz Used and Notes ( )-N-42-hydroxy-4,6- 11 NMR (300 MHz, CD:30D):
dimethylpyridin-3- ö 8.22-8.13 (m, 2H), 7.28-7.15 146 A. ft C yl)methyl)-2-methyl-1-(1- (m, 611), 6.47-6.45 (m, 1H), phenylethyl)-1H- 6.09 (s, 1H), 4.50 (s, 214), 2.42 pyrrolo[2,3-bipyridine-3- (s, 3H), 2.39 (s, 3H), 2.23 (s, carboxami de 3H), 2.02 (d, .1= 7.2 Hz, 3H).

General Methods Compound Name NMR data m/z Used and Notes 1H NMR (300 MHz, tt-DMS0): 6 11.57 (s, 1H), 8.13 1-benzoyl-N-((2-hydroxy- (q, J= 5.1 Hz,1H), 7.74-7.70 150 A. B, C 4,6-dimethylpyridin-3- (m, 411), 7.60-7.58 (m, 2H), yl)methyl)-2-methyl-1H- 7.20-7.00 (m, 3H), 5.88 (S, indole-3-carboxamide 1H), 4.32 (d, J= 5.1 Hz, 2H), 2.37 (S, 3H), 2.267 (S, 3H), 2.12 (S, 3H).
IH.NMR (300 MHz, CD30D):
N-((4,6-dimethy1-2-oxo- 6 9.03 (s, 1H), 8.53 1,2-dihydropyridin-3- .. (s,2H),7.80-7.78 (m, 1H), A, , yl)methyl)-2-methyl-1- 7.39-7.36 (m, 1H), 7.19-7.16 401 (pyrimidin-5-ylmethyl)- (m, 2H), 5.56 (s, 2E1), 4.55 (s, 1H-indole-3-carboxamide 2H), 2.62 (s, 3H), 2.42 (s, 3H), 2.45 (s, 3H).
NMR (300 MHz, CD30D):
7.73-7.70 (m, 1I1), 7.56-7.53 (R or S)-1-(sec-butyl)-N- (m 1 II), 7.12-7.09 (m, 2H), 6- ((2-hydroxy-4, 147 A, B, C, D 6.11 (s, 111), 4.54 (s, MP, 2.64 dimethy1pyridin-3-yl)meth y 'l)-2-methyl-1 H-(s, 3H), 2.43 (s, 31-1), 2.25 (s, J65 3H) 1 97-1 90 (m /1-1) 1.61 indole-3-carboxamide ' ' ¨ ' (d, J= 6.9 Hz, 3110, 0.73 (t, J
7.5 Hz, 3H).
(R or S)-1-(sec-buty1)-N-148 A B C D ((2-hydroxy-4,6-, , , dimethylpyridin-3- Identical to Compound 147 365 yOmethyl)-2-methyl-1H-indolc-3-carboxamide (R)-2-chloro-N-((2-hydroxy-4,6-Starting material dimethylpyridin-3-167 for other yOmethyl)-6-methyl-7-(1-pyrrolo[2,3-phenylethyl)-7H-cljpyrimidines pyrrolo[2,3-cl]pyrimidine-5-carboxamide J1T NMR (400 MHz. CD30D) (R)-N-((2-hydroxy-4,6-6 8.95 (s, 11-1), 7.29-7.23 (m, dimethylpyridin-3-5H), 6.20-6.18 (m, 1H), 6.09 yl)methyl)-6-methyl-7-(1-(s, 1 11), 4.49 (s, 168 F. G phenylethyl)-2-3.99 (m, 211), 3.59-3.53 (m, 2 (tetrahydro-2H-man-4-1-1), 3.17-3.02 (m, 1 H), 2.55 y1)-7H-pyrrolo[2,3-(s, 3H), 2.38 (s, 3H), 2.22 (s, dipyrimidine- ../=

3H), 2.09 (d, .7.2 Hz, 311), carboxamide 1.96-1.88 (m, 4H).

General Methods Compound Name NMR data m/z Used and Notes (R)-N-((2-hydroxy-4,6- 'H NMR (400 MHz, CD30D) dimethylpyridin-3- 6 8.64 (s, 1H), 7.30-7.21 (m, 169 E yl)methyl)-6-methy1-7-(1- 5H), 6.09-6.04 (m, 2H), 4.47 phenylethyl)-2-(piperazin- (s, 2 H), 3.72-3.67 (m, 4H), 1-y1)-7H-pyrrolo[2,3- 2.84-2.81 (m, 4 H), 2.44 (s, d]pyrimidine-5- 311), 2.37 (s, 3H), 2.23 (s, 3H), carboxamide 2.01 (d, J=7.2 Hz, 311).
111 NMR (400 MHz, CD30D) (R)-N-((2-hydroxy-4,6- 8.93 (s, 1H), 7.30-7.18 (m, dimethylpyridin-3- 5H), 6.42-6.35 (m, 1H), 6.09 176 11, B. C yl)methyl)-2,6-dimethyl- (s, 1 H), 4.48 (s, 2 H), 2.69 (s, 7-(1-phenylethyl)-7H- 3H), 2.42 (s, 3 H), 2.37 (s, 3 pyrrolo[2,3-d]pyrimidine- H), 2.22 (s, 311), 2.04 (d, 5-carboxamide J=7.2 Hz 311).
NMR (400 MHz, (R)-N-((2-hydroxy-4,6- Methanol-d4) (58.92 (s, III).
dimethylpyridin-3- 7.29-7.22 (m, 5H), 6.21-6.09 177 F, G, B, C yl)methyl)-2-isopropyl-6- (m, 211), 4.49 (s, 2 II), 3.25-methy1-7-(1-phenylethyl)- 3.22 (m, 1 H), 2.53 (s, 3H), 7H-pyrrolo[2,3- 2.38 (s, 3 H), 2.23 (d, J= 0.4, dipyrimidine-5- 3 H), 2.09 (d, J=8.0, 3 H), carboxamide 1.33-1.26 (m, 611).
111 NMR (400 MHz, CDC13):
(R)-7-(sec-buty1)-2-8.94 (s, 1H), 6.85 (s, 1.H), chloro-N-04,6-dimethyl-4.62 (s, 2H), 2.72 (s, 3H), 2.59 184 See Intermediate 2-oxo-1,2-dihydropyridin-1 synthesis below 3-yl)methyl)-6-methyl- (s, .511), -.4.5 (s, 3H), z35-2.41 (m, 1H), 1.94-2.04 (m, 7H-pyrrolo[2,3-1H), 1.65-1.67 (d, J= 7.2 Hz, dipyrimidine-5-3H), 0.72-0.76 (t, 3H).
carboxamide 1H NMR (400 MHz, (R)-7-(sec-buty1)-N-02-Methanol-d4) 69.57 (s, hydroxy-4,6-1H),9.40 (s, 111), 8.50 (s, 2H).
dimethylpyridin-3-1 88 7.53 (s, 3 H), 6.48 (sõ
yl)methyl)-6-methyl-2-1H),4.56-4.65 (m, 3H), 2.86(s, pheny1-711-pyrrolo[2,3-311), 2.63 (s, 3 H), 2.55 (s, 3 d]pyrimidine-5-H), 2.41 (s, 1 H), 2.10 (s, 1 H), carboxamide 1.78 (s, 311), 0.83 (s, 311).

General Methods Compound Name NMR data m/z Used and Notes 3H NMR (400 MHz, CD301)):
(R)-7-(sec-buty1)-N-02- ô 9.4 (s, 1H), 9.14 (s, 1H), hydroxy-4,6- 6.86 (s, 1H), 4.79-4.83 (m, 189 G dimethylpyridin-3- 1H), 4.65 (s, 2H), 2.82 (s, 3H), 368 yOmethyl)-6-methyl-7H- 2.62 (s, 3H), 2.48 (s, 3H), pyrrolo[2,3-d]pyrimidine- 2.39-2.44 (m, 1H), 2.05-2.12 5-carboxamide (m, 1H), 1.72-1.74 (d, J = 6.8 Hz, 3H), 0.77-0.81 (t, 3H).
NMR (400 MHz, (R)-7-(sec-butyl)-N-((4,6- Methanol-d4) (59.20 (s, 1H), dimethy1-2-oxo-1,2-6.64 (s, 111), 4.73 (s, 214), 4.59 dihydropyridin-3-197 F. G (s, 211), 3.44 (m, 111), 2.77 (s, yOmethyl)-2-isopropyl-6- 3H), 2.55 (s, 3 H), 2.4 (s, methy1-711-pyrrolo[2,3-3H), 2.10 (m, 1H), 1.73(d, I =
d)pyrimidine-5-6.8Hz,3 H), 1.48(d, J=
carboxamide 7.2Hz,6H)õØ81 Lm, (R)-7-(see-buty1)-N5- NMR (400 MHz, F (Zn(CN)2 ((4,6-dimethy1-2-oxo-1,2- Methanol-d4) 69.34 (s, 1H), instead of the dihydropyridin-3- 6.4 (s, 11-1), 4.92 (s, 211), 4.58 .
Indicated boronic yl)methyl)-N2,6- (s, 21-1), 3.1 (s, 311), 2.86(s, 425 ester or acid), 1, C dimethy1-7H-pyrrolo[2,3- 31-I), 2.50 (s, 3H), 2.34(s, 311), d]pyrimidine-2,5- 1.80 (d, J=6.8 I1z,3 H), 0.86 dicarboxamide (m, 311).
(R)-7-(sec-buty1)-N-((4,6- 11-1NMR (400 MHz, dimethy1-2-oxo-1,2- Methanol-d4) (58.97 (s, 1H), F (Zn dihydropyridin-3- 6.1 (s, 1H), 4.76 (s, 2H), 4.50 (CN )2), 1, .1, 199 yl)methyl)-2- (s, 2H), 2.67 (s, 3H), 2.51(s, 398 (hydroxymethyl.)-6- 1H), 2.44 (s, 310, 2.42(s, 311), methyl-7H-pyrrolo[2,3- 2.01 (m, 2 H), 1.95(d, d]pyrimidine-5- J=6.8Hz, 3H), 0.74 (m, 3H).
carboxamide (R)-7-(sec-butyl)-N-((4,6- 1H NMR (400 MHz, dimethy1-2-oxo-1,2- Methanol-d4) (58.87 (s, 1H), 200 F dihydropyridin-3- 6.09 (s, 1H), 4.71 (s, 1H), 4.62 yOmethyl)-2,6-dimethyl- (s, 2H), 2.67 (d, I =6.4Hz, 7H-pyrrolo[2,3- 6H), 2.51 (m, 4H), 2.23 (s, 3 d)pyrimidine-5- 1 H), 2.00 (m, 1H), 1.65(d, .1 carboxamide =6.8 Hz, 3 H), 0.73 (m, 311).

General Methods Compound Name NMR data m/z Used and Notes (R)-2-amino-N-((2- 3H NMR (300 MHz, CD30D):
hydroxy-4,6- ö 8.57 (s, 1H), 7.34-7.19 (m, 144 dimethylpyridin-3- 5H), 6.19-6.17 (m, 1H), 6.10 yl)methyl)-6-methyl-7-(1- (s, 1H), 4.47 (s, 2H), 2.33 (s, 430 phenylethyl)-7H- 3H), 2.30 (s, 3H), 2.23 (s, 3H), pyrrolo[2,3-d]pyrimidine- 1.99 (d, J= 7.2 Hz, 3H).
5-carboxamide (R)-N-42-hydroxy-4,6- 111 NMR (400 MHz, CD30D) dimethylpyridin-3- 8 8.56 (s, 1H), 7.27 7.20 (m, yl)methyl)-6-methy1-7-(1- 5H), 6.06 ¨ 6.02 (m, 2H), 4.45 phenylethyl)-2- (s, 211), 3.52 3.45 (in, 410, 484 (pyrrolidin-1 -y1)-7H- 2.40 (s, 3H), 2.35 (s, 311), 2.21 pyffolo[2,3-d]pyrimidine- (s, 3H), 2.01 ¨ 1.94 (m, 7H).
5-carboxamide (R)-N-((2-hydroxy-4,6-NMR (400 MHz, CD30D) 6 8.65 (s, 1H), 7.28 (dt, J =
dimethylpyridin-3-11.9, 7.7 Hz, 5H), 6.10 (dd, J
143 yl)methy1)-6-methyl-2-= 13.7, 6.5 Hz, 2H), 4.51 (s, mmpholino-7-(1- 500 2H), 3.78 ¨3.65 (m, 8H), 2.47 phenylethyl)-7H-(s, 3H), 2.41 (s, 3H) 2.26 (s.
pyrrolo[2,3-d]pyrimidine- ' 311), 2.03 (d, J = 7.2 Hz, 311).
5-carboxamide (R)-N-((2-hydroxy-4,6-1H NMR (400 MHz, CD30D) dimethylpyridin-3-8 8.75 (s, 1I1), 7.29 ¨ 7.22 (m, 141 (Na0MeiMe0H yl)methyl)-2-methoxy-6-5H), 6.11 ¨6.07 (m 2H) 4.46 methy1-7-(1-phenylethyl)-" 445 instead of amine) 7 (s, 21I), 3.90 (s, 31I), 2.48 (s, 11-pyrrolo[2, 3-3H), 2.36 (s, 3H), 2.21 (s, 3H), d]pyrimidine-5-2.05 (d, J= 7.2 Hz, 3H).
carboxamide 'H NMR (400 MHz, CD30D):
( )-N((4,6-dimethy1-2- 6 7.22-7.13 (in, 411), 7.01 (d, J
oxo-1,2-dihydropyridin-3- = 7.2 Hz, 2H) ,6.69 (d, J =

yl)methyl)-2,6-dimethyl- 7.2 Hz, 1H), 5.99 (s, 1H), 4.35 A, , 7-oxo-1-(1-phenylethy1)- (s, 2I1), 3.50-3.21 (m, 4I1), 444 6,7-dihydro-1H- 2.26 (s, 3H), 2.13 (s, 311), 2.06 pyrrolo[2,3-c]pyridine-3- (s, 311), 1.82 (dõ/ = 7.2 Hz, carboxamide 311).

General Methods Compound Name NMR data m/z Used and Notes (R)-N-02-hydroxy-4,6- 1H NMR (300 MHz, CD3011):
dimethylpyridin-3-o 8.55 (s, 111), 7.30-7.23 (m, 140 yOmethyl)-6-methyl-2-5H), 6.15-6.10 (m, 2H), 4.47 111 (methylatnino)-7-(1-(s, 2H), 3.25 (s, 3H), 2.40 (s, phenylethyl)-7H-31), 2.38 (s, 3H), 2.24 (s, 311), pyrrolo[2,3-d]pyrimidine-2.03 (d, J = 7.2 Hz, 3H).
5-carboxamide 1H NMR (300 MHz, CDC13) 5 12.62 (s, 1H), 7.89 (dõ I= 8.0 N-((2-hydroxy-4,6-Hz, I H), 7.77 7.40 (m, 3H), 133 dimethylpyridin-3-B, C 7.43 7.18 (m, 3H), 7.22 --yl)methyl)-2-methyl-1- 385 6.94 (m, 2H), 5.98 (s, 1H), pheny I-1H-i ndo I e-3-4.65 (s, 2H), 2.60 ¨ 2.52 (m, carboxamide 3H), 2.51 ¨ 2.44 (m, 3H), 2.28 ¨ 2.22 (m, 31).
111 NMR (300 MHz, d6-DMS0): 6 11.61 (s, 1.11), 7.80-7.69 (m, 211), 7.46-7.42 (m, 1-benzyl-N-((2-hydroxy-1H), 7.30-7.22 (m, 311), 7.11-119 A. B. C 4,6-dimethylpyridin-3-7.07 (m, 2H), 6.98 (d, J= 7.5 399 yl)methyl)-2-methyl-111-Hz, 211), 5.89 (s, 111), 5.46 (s, indole-3-carboxamide 211), 4.33 (d, J= 4.5 Hz, 211), 2.57 (s, 311), 2.26 (s, 31{), 2.11 (s, 3H).
111 NMR (300 MHz, d5-DMS0) (311.46 (s, 1H), 9.49 (t, Ji = 4.2 Hz, J2=8.1 Hz, ( )-N4(2-hydroxy-4,6-1H), 8.28 (d, J= 3.6 Hz, 1H), dimethylpyridin-3-7.50 (d, J= 6.3 Hz, I H), 7.36-126 B, C yl)methyl)-2-methy1-1-(1-7.27 (m, 3H), 7.16 (d, J= 5.7 415 phenylethyl)- I H-Hz, 1H), 7.03-7.00 (m, 1H), pyrrolo[3,2-b]pyridine-3-6.03 (m, 1H), 5.85 (s, 1H), carboxamide 4.37 (s, 211), 2.89 (s, 3H), 2.10 (s, 3H), 1.89 (d, J= 5.4 Hz, 1F1).

General Methods Compound Name NMR data m/z Used and Notes 1H NMR (300 MHz, tt-DMS0): 6 11.59 (s, 1H), 7.75-(R or S)-N-((6-hydroxy-2- 7.72 (m, 2H), 7.35-7.25 (m, methoxy-4- 3H), 7.15 (dõI = 7.5 Hz, 2H), 114 A, B, C, D methylpyridin-3- 7.10-7.07 (m, 1H), 7.02-6.93 yOmethyl)-2-methyl-1-(1- (m, 2H), 6.15 (s, 1H), 5.96 (q, phenylethyl)-1H-indole-3- J= 6.9 Hz, 111), 4.33 (d, J =
carboxamide 5.1 Hz, 2H), 3.85 (s, 3H), 2.62 (s, 3H), 2.20 (s, 3H), 1.89 (d, J
= 7.5 Hz, 3H).
(R or S)-N-((6-hydroxy-2-methoxy-4-115 A, B, C, D methylpyridin-3- Identical to Comopund 114 yOmethyl)-2-methyl-1-(1-phenylethyl)-1H-indole-3-i carboxamide NMR (300 MHzõ -DMS0): (3. 11.50 (s, 1H), ( )-N-42-hydroxy-4,6- 8.46 (s, 111), 8.16-8.13 (m, 118 A B dimethylpyridin-3- 1H), 7.81 (s, 1H), 7.42-7.39 ., , C
yl)methyl)-1-(1- (m, 111), 7.31-7.20 (m, 511), phenylethyl)-1H-indole-3- 7.10-7.07 (m, 2H), 5.88-5.75 carboxamide (m, 211), 4.32 (d, = 5.1 Hz, 2H), 2.22 (s, 3H), 2.12 (s, 3H), 1.86 (d, I = 6.9 Hz, 3H).
IFINMR (DMSO-d6, 300 MHz) 6 11.59 (s, 1H), 7.73-(R or S)-N-((2-hydroxy- 7.66 (m, 2H), 7.34-7.29 (m, 137 A B D 4,6-dimethylpyridin-3- 3H), 7.24 (d, J= 7.2 Hz, 1H), , , C, yl)methyl)-2-methy1-1-(1- 7.16-6.89 (m, 4H), 5.94 (q, 413 phenylethyl)-1H-indole-3- 1H), 5.88 (s, 1H), 4.32 (d, J=
carboxamide 5.4 Hz, 2H), 2.60 (s, 3H), 2.50 (s, 311), 2.11(s, 3H), 1.87 (d, J
= 7.2 Hz, 311).
(R or S)-N-((2-hydroxy-A B D
136 4,6-dimethylpyridin-3-, , C, Amethyl)-2-methyl-1-(1- Identical to Compound 137 phenylethyl)-1H-indole-3-carboxamide General Methods Compound Name NMR data m/z Used and Notes 1H NMR (400 MHz, ce-DMS0): 6 11.58 (s, IH), 8.50 (s, 1H), 8.21 (d, J= 8.4 Hz, (R or S)-N-((2-hydroxy- 1H), 7.89 (s, 1H), 7.73 (s, IH), B C D 4,6-dimethylpyridin-3- 7.64 (d, J= 7.2 Hz, 2H), 7.46-, , yl)methyl)-2-phenyl-1-(1- 7.42 (m., 3H), 7.33-7.27 (m, 475 phenylethyl)-1H-indole-3- 5H), 7.25-7.21(m, 1H), 6.05-carboxamide 6.02 (m, 1H), 5.91 (s, 1H), 4.34-4.33 (d, J = 3.2 Hz, 2H), 2.42 (s, 3H), 2.13 (s, 3H), 1.89-1.88 (d, J = 7.2 Hz, 311).
1H NMR (400 MHz, d6-DMS0): 6 11.54(s, IH), 8.51 (R or S)-N-((2-hydroxy- (s, 1H), 8.42 (s, 1H), 7.91 (s, 446 B C D 4,6-dimethylpyridin-3- 1H), 7.63 (d, .1= 7.2 Hz, 211), , , yl)methyl)-2-phenyl-1-(1- 7.51-7.40 (m, 41-1), 7.33-7.24 475 phenylethyl)-1H-indole-3- (m, 6H), 5.90 (m, 2H), 4.35-carboxamide 4.34 (d, .1= 3.6 Hz, 2H), 2.34 (s, 3H), 2.13 (s, 3H), 1.89-1.87 (dõi= 6.8 Hz, 3H).
111 NMR (300 MHz, CD30D,): 6 8.06(s, 1H), 7.92 ( )-2-cyclopropyl-N-02- (d, J = 8.4 Hz, 1H), 7.28-7.15 hydroxy-4,6- (m, 5H), 7.00 (s, 1H), 6.87 (d, 131 B, C dimethylpyridin-3- J = 8.1 Hz, 1H), 6.13(s, 1H), 43, yl)methyl)-1-(1- 5.74 (q, J-7.2 Hz, 111), 4.52 phenylethyl)-1H-indole-3- (s, 2H), 2.41 (s, 3H), 2.25 (s, carboxamide 3H), 1.91 (d, .1= 7.2 Hz, 3H), 1.93-1.89 (m, 114), 0.92-0.88 __________________________________________ r-(m, 2H), 0.61-0.59 (m, 2H).
.1-1H NMR (400 MHz, d6-DMS0): 6 11.58 (s, 1H), 8.37 (- )-2-ethyl-N42- (s, 1H), 8.03 (d, J= 8.4 Hz, hydroxy-4,6- 1H), 7.80 (s, 1H), 7.32-7.20 447 B, C dimethylpyridin-3- (m, 611), 6.98-6.96 (m, 1H), 41.7 yl)methyl)- I -(1- 5.91(s, 111), 5.85-5.80 (m, phenylethyl)-1H-indole-3- 1H), 4.31 (s, 2171), 2.66-2.60 carboxamide (m, 2H), 2.23 (s, 311), 2.13 (s, 3H), 1.85-1.83 (d, J = 7.2 Hz, 311), 1.21-1.14 (m, 3H).

General Methods Compound Name NMR data m/z Used and Notes NMR (400 MHz, Me0D):
(S)-N-((4-methoxy-6- ô 9.311 (S, 1H),9.122(S, 1H),1 methyl-2-oxo-1,2- 6.826 (S, 1H), 5.063-5.027 dihydropyridin-3- (m, 1H), 4.578 (s, 2H), 4.279-See intermeidate 287 yOmethyl)-7-(1- 4.254 (t, J=10 Hz.1H), 4.086 400 1 below, G
methoxypropan-2-y1)-6- (s,3H) ,3.781-3.744 (m, 1H), methyl-7H-pyrrolo[2,3- 3.203 (s, 3H), 2.805 (s, 3H), dipyrimidine-5- 2.500 (s, 3H), 1.739-1. 722 (d, carboxamide J= 6.8 Hz, 3H).
11-1 NMR (400 MHz, Me0D):
(R)-N-((4-methoxy-6-(5 9.352 (S, 1H),9.152(S, 1H), methy1-2-oxo-1,2-7.018 (S, 1H), 5.075-4.994 dihydropyridin-3-See intermediate (m, 1H), 4.610 (s, 2H), 4.280-303 yl)methyl)-7-(1-1 below, G 4.230 (t, J= 20 Hz,1H), 4.139 400 methoxypropan-2-y1)-6-(s,3H) ,3.784-3.747 (m, 1H), methy1-7H-pyrrolo[2,3-3./04 (s, 3H), 2.806 (s, 3H), dlpyrimidine-5-1.567 (s, 3H), 1.748-1. 730 (d, carboxamide J = 7.2Hz, 3H).
111 NMR (CDC13, 400 M Hz) S 11.99(s, 1H), 9.17 (s, 1H), 8.77 (s, 1H), 7.61 (s, 1H), 5.96 (s, 1H), 4.61 (d, J = 4.0 Hz, 2H), 4.04 (dd, Jj = 3.6 Hz, //
(R or S)-N-((4-methoxy-11.2 Hz, 1H), 3.91 (s, 3H), 6-methy1-2-oxo-1,2-3.76 (dd, Ji ---- 3.6 Hz, .12 dihydropyridin-3-See intermediate 11.2 Hz, 1H), 3.43 (t, J=
3 19 yl)methy1)-6-methy1-741-8 below, G, B, C 11.2Hz, 1H), 3.18 (dd, ././

(tetrahydro-2H-pyran-4- .
10.4 Hz, 12 = 12.0 Hz, 1H), ypethyl)-7H-pyrrolo[2,3-1.85 (s, 1H), 1.77 (s, 3H), 2.30 dipyrimidine-5-(s, 3H), 1.89 (d, .1¨ 12.0 Hz, carboxamide 1H), 1.64 (d, J= 6.4 Hz, 41]), 1.40 (d, J= 8.4 Hz, 1H), 1.13 (dd,J1= 4.8Hz, .1.2 =12.8 Hz, 1H), 1.10-0.79(m., 1H).

General Methods Compound Name NMR data tniz Used and Notes 'H. NMR (400 MHz, CD3C13):
8 0.93 (m, 2H), 1.24 (m, 111), tert-butyl 4-(1-(5-(((4- 1.42 (s, 9H), 1.67 (m, 6H), methoxy-6-methyl-2-oxo- 1.96-1.99 (d, J=12.4Hz, 1H), 1,2-dihydropyridin-3- 2.30 (s, 3H), 2.48 (m. 1H), intermediate 9, B, 332 yl)methypearbamoy1)-6- 2.78 (s, 4H), 3.9 (s, 4H), methyl-7H-pyrrolo[2,3- 4.20(brs, 1H), 4.65-4.66(d.
d]pyrimidin-7- J=4.0Hz, yl)ethyl)piperidine-1- 2110,5.96(s,1H),7.71(brs,1 H), carboxylate 8.78(s, 11-1), 9.19 ( s, 111) ,12.67(brs, 1H).
( )-N-((4-methoxy-6- 'H NMR (400 MHz, CDC13):
methyl-2-oxo-1,2- 8 1.58 (d, J=6.4 Hz, 6H), 2.54 IFA treatment of dihydropyridin-3- (s, 3H), 3.94 (s, 3H), 4.66 (d, Compound 332 yl)methyl)-6-methyl-7-(1- J=5.6 Hz, 2H), 4.78(m, 1H), (piperidin-4-ypethyl)-7H- 6.56 (brs, 111), 6.71 (s, 1H), pyrrolo[2,3-d]pyrimidine- 7.54 (s, 1H), 7.73 (s, 1H), 5-carboxamide 8.27 (s, 1F1), 8.37 (s, 1171), 111 NMR (CDC13, 400MHz) 8 ( )-7-(1-(4,4- 9.19 (s, 1 Fl), 8.79 (s , 111), Synthesis for difluorocyclohexyl)ethyl)- 7.69 (s, 1 fi) , 5.96 (s, 1H), intermediate 9, N-((4-methoxy-6-methyl- 4.65-4.64 (d ,J=7.2Hz , 211), 334 using 2-oxo-1,2-dihydropyridin- 3.91 (s , 311), 3.02 ( s , 4H), intermediate 10, 3-yl)methyl)-6-methyl- 2.78 (s , 311), 2.31 (s , 311), B, C. 7H-pyrrolo[2,3- 2.22-2.07 (m , 311), 1.89 (s, d]pyrimidine-5- 2H), 1.69 (s 3H) , 1.48-1.42 carboxamide (m , 2H) , 1.14 (s , 3H), 1.11-1.01 (rn , 2H).
1H NMR (CDC13, 400MHz) 8 9.23-9.22 (d, J= 0.8 Hz, 1H), 9.17-9.16 (d, J= 1.2Hz, 1H), 6.56 (s, 111), 4.98-4.94 (m, Synthesis for N-((4-methoxy-6-methvl-- 1H), 4.76-4.72 (m, 1H), 4.55 intermediate 9 2-oxo-1 ,2-dihydropyridin-using 3-yl)methyl)-6-methyl-7- (s, 211), 4.27-4.23 (dd, J=

intermediate 11, (1((R)-mmpholin-2- 3.6 Hz, 7.2Hz, 1H), 4.03 (s, 4.4.1 G, BOC-on, B, C, ypethyl)-7H-pyrrolor,3- 31), 4.00-3.96 (m, 1H), 3.40-TFA dipyrimidine-5- 3.37 (d, J¨ 13.2 Hzõ 1H), carboxamide 3.26-3.16 (m, 2H), 3.10-3.04 (m, 111), 2.78 (s, 311), 2.44 (s, 3H), 1.83-1.85 (d, J= 7.2 Hz, 3H).

General Methods Compound Name NMR data m/z Used and Notes JH NMR (CDC13, 400 M Hz) 12.12 (s, 1H), 9.20 (s, 1H), 8.78 (s, 1H), 7.68 (s, 1H), 5.97 (s, 1H), 4.70-4.58 (m, 2H), 3.91 (s, 4H), 3.62 (d, J= 12.0 General Hz, 1H), 2.76-2.67 (m, 711), 370 1 2.45 (dd. Ji = 2.0 Hz, .12 =
procedure L on 517 1 11.6 Hz, 1H), 2.32 (s, 3H), Compound 333 2.10 (t, J= 12.0 Hz, 1H), 1.67 (d, J = 6.8 Hz, 4H), 1.48-1.40 (m, Ili), 1.37-1.29 (m, 1H), 1.27-1.19 (m, 111), 0.9-0.78 (m, 1H).

Example 73. Synthesis of (R)-N-02-hYdroxv-4,6-dimethylpyridin-3-yftmethyl)-6-methyl-7-(1-phenylethyl)-711-pyrroloi2,3-dlpyrimidine-5-carboxamide (Compund 395). To a solution of (R)-2-ch loro-N42-hydroxy -4,6-dimethylpyri din- 3-yl)methyl)-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-di pyrimidine-5- carboxamide (140 mg, 0.30 mmol) in Me0H (10 mL), Palladium-carbon catalyst (10%, 20 mg) was added, the mixture solution was stirred at 25 C for 12 hours under hydrogen atmosphere (4 bar). The mixture was filtered and concentrated in vacuo and purified by column chromatography (silica gel, dichloromethane/methanol =
10: 1) to afford (R)-N-((2-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-7-(1-phenylethyl)-pyrrolo[2,3-d]pyrimidine-5-carboxamide (93 mg, 72%).
005271 Example 74. Synthesis of novel indole cores and intermediates.
[005281 Intermediate 1: Intermediate 1 was synthesized according to the scheme below:
N?"1Sr Br NH2 0 N -'---,X. 4 0 ..T.4...,µ DIPEA, Et0H (et N'' NH ji......
Pd(OAc)2. L CL K2CO2. UHF
__________________________ r. . ____________________ ii.
,x,/, -, y Step 1 $tep 2 O\_1 / 0.....
...:6-CM :
..*¨ kle011 1 HI' -g.. .\-- i EDC! ROM: et2N
,,IL L=-,,, ----L-- A , Step 3 Ci NI N
d Oil [00529] Step 1: (R)-5-bromo-2-chloro-N-(1-phenylethyl)primidin-4-amine. To a solution of 5-bromo-2,4-dichloropyrimidine (5 g, 22 mmol) and (R)-1-phenylethanamine (2.7 g, 22 mmol) in ethanol (50 mL) was added N,N-diisopropylethylamine (4.3 g, 33 mmol). The reaction solution was stirred at room temperature for 12 hours. The resultant mixture was concentrated in vacuo. The residue was purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 6:1) to give (R)-5-bromo-2-chloro-N-(1-phenylethyl)pyrimidin-4-amine as a white solid (5 g, 74%). LRMS (M + 11') m/z: calcd 310.98; found 310. 'H NMR
(300 MHz, CDC13): 6 8.10 (s, 1H), 7.41-7.25 (m, 5H), 5.73 (d, J = 6.9 Hz, 1H), 5.39-5.34 (m, 1H), 1.61 (d, J= 6.6 Hz, 3H).
1005301 Step 2: (R)-methyl 2-chIoro-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-dlpyrimidine- 5-carboxylate. A solution of (R)-5-bromo-2-ehloro-N-(1-phenylethyl)pyrimidin-4-amine (5 g, 16 mmol), methyl but-2-ynoate (3.1 g, 32 mmol), lithium chloride (690 mg, 16 mmol), potassium carbonate (5.5 g, 40 mmol) and palladium acetate (360 mg, 1.6 mmol) in N,N-dimethylformamide (50 mL) was degassed and back-filled with nitrogen for three times, then heated at 120 C for 4 hours. The reaction mixture was filtered and the filtrate was concentrated in vacuo, extracted with ethyl acetate (50 mL), washed with water (50 mL), dried over anhydrous magnesium sulfate and purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 5:1) to give (R)-methyl 2-chloro-6-methy1-7-(l-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylate as a yellow oil (800 mg, 15%). LRMS (M + H+) m/z:
calcd 329.09;
found 329. 'H NMR (300 MHz, CDC13): 6 9.16 (s, 1H), 7.39-7.18 (m, 5H), 6.45-6.42 (m, 1H), 3.96 (s, 3H), 2.57 (s, 3H), 2.05 (d, .1= 7.2 Hz, 3H).
1005311 Step 3: (R)-2-chloro-6-methy1-7-(1-phenylethyl)-7H-pyrrolo [2,3 -dipyrimi dine-5-carboxylic acid. Lithium hydroxide anhydrate (882 mg, 21 mmol) in water (3 mL) was added to (10-methyl 2-chloro-6-methy1-7-(1-phenylethyp-7H-pyrrolo[2,3-dlpyrirnidine-5-carboxylate (700 mg, 2.1 mmol) in tetrahydrofuran (5 mL) and methanol (10 mL) and the resultant mixture was stirred at room temperature for 12 hours. The mixture was evaporated, added with water (5 mL), acidified with aqueous hydrochloric acid (1M) to pH = 2. The precipitate solid was filtered and dried to obtain (R)-2-chloro-6-methy1-7-(1-phenylethyl)-7H-pyrrolo[2,3-dj pyrimidine-5-carboxylic acid as a white solid (500 mg, 75%). LRMS (M It) m/z: calcd 315.08;
found 315.
[00532] Step 4: (R)-2-chloro-N42-hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-7- (1-phenylethy1)-7H-pyrrolo12,3-dipyrimidine-5-carboxamide. To a solution of (R)-2-chloro-6-methy1-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylic acid (100 mg , 0.32 mmol) in dichloromethane (10 mL) was added with 1-hydroxybenzoniazole (65 mg, 0.48 mmol), (3-dimethylaminopropyl)ethyl-carbodiimid hydrochloride (92 mg, 0.48 mmol) and triethylamine (97 mg, 0.96 mmol). After stirred for 30 minutes, 3-(aminomethy1)-4,6-dimethylpyridin-2-ol (49 mg, 0.32 mmol) was added and the reaction mixture was stirred at room temperature for 12 hours. The solution was concentrated, diluted with water (20 mL), extracted with ethyl acetate (20 mL). The organic layers were separated, combined, dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 20: 1) to afford (R)-2-eh loro-N4(2- hydroxy-4,6-dimethylpyridin-3-yl)methyl)-6-methyl-7-(1-phenylethyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxamide (100 mg, 70%). LRMS (M + 1.14) calcd 449.16;
found 449.
[005331 Intermediate 2. Intermediate 2 was synthesized according to the scheme below:

(1:r NvcI0 )L), N -0 toei/NaH/Oke 0 9 Na rH/THF NO2 -a NH4Cl/EtOli - No2 Stepl NO2 Step2 Step3 o o I
[005341 Step I: 4-chloro-1-methyl-3-nitropyridin-2(IH)-one. To a stirred solution of 4-ch1oro-3-nitropyridin-2(1f1)-one (3.0 g, 17 mmol) in N,N-dimethylformamide (50 mL) was added sodium hydride (60% w/w, 1.0 g, 25.5 mmol) in batches at 0 C. The mixture was stirred at room temperature for 30 minutes. Then iodomethane (2.9 g, 20.4 mmol) was added dropwise to the above solution at room temperature .The resultant solution was stirred at room temperature for 12 hours. Once starting material was consumed, the reaction mixture was quenched with ice water (100 mL) at 0-10 C, and extracted with ethyl acetate(100 mL * 3).The organic phase was washed with brine (100 mL * 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 10:1) to give 4-chloro- 1 -methyl-3- nitropyridin-2(1H)-one (3 g, 94%) as a yellow solid. LRMS (M + 11+) mtz: calcd 188.0; found 188.
[005351 Step 2: ethyl 2-(1-methyl-3-nitro-2-oxo-1,2-dihydropyridin-4-yI)-3-oxobutanoate. To a stirred solution of ethyl 3-oxobutanoate (2.5 g, 19 mmol) in tetrahydrofuran (50 mL) was added sodium hydride (60% w/w, 0.96 g, 23.9 mmol) in batches at 0 C. The mixture was stirred at room temperature for 30 minutes .Then the solution of 4-chloro-1-methy1-3-nitropyridin-2(1H)-one (3.0 g, 16 mmol) in tetrahydrofuran (50 mL) was added in one portion.
The resultant solution was stirred and heated to 50 C for 12 hours. Once starting material was consumed, the reaction solution was quenched with water (100 mL) at 0 C, and extracted with ethyl acetate (100 mL * 3). The combined organic layers were washed with brine (100 mL * 2), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethanelmethanol =
10:1) to give ethyl 2-(1-methy1-3-nitro-2-oxo-1,2-dihydropyridin-4-y1)-3-oxobutanoate (2.5 g, 56%) as a yellow solid. LRMS (M + Fr) m/z: calcd 282.09; found 282.
1005361 Step 3: ethyl 2,6-dirnethy1-7-oxo-6,7-dihydro4H-pyrrolo[2,3-cjpyridine-carboxylate. To a solution of ethyl 2-(1-methy1-3-nitro-2-oxo-1,2-dihydropyridin-4-y1)-3-oxobutanoate (2.5 g, 8.8 mmol) in ethanol (50 mL) was added ammonium chloride (0.5 g, 9 mmol) in water (5 mL) at room temperature .The mixture was stirred and heated to reflux. Then iron powder (0.5 g, 8.9 mmol) was added in one portion. The mixture was stirred at reflux for 2 hours. Once starting material was consumed, the resultant mixture was filtered when it was hot, and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (silica gel, dichloromethane/methanol = 10:1) to give ethyl 2,6-dimethy1-7-oxo-6,7-dihydro-IH-pyrrolo [2,3-c]pyridine-3-carboxylate (1.5 g, 75%) as a brown solid. LRMS (M
+ Fr) m/z: calcd 234.1; found 234. III NMR (400 MHz, da-DMS0): (5 12.54 (s, 1H), 7.30 (d, 5.1 Hz, 111), 6.78 (d, J = 5.1 Hz, 111), 4.24 (q, j:: 5.1 Hz, 211), 3.51 (s, 3H), 2.56 (s, 314), 1.32 (t, 5.1 Hz, 3H).
l005371 Intermediate 3. Intermediate 3 was synthesized according to the scheme below:
HO, OH
13- Cu(OAc)2. 4A MS

0(j DMAP, Et3N ' 0 0 step 1 0 A mixture of methyl 2-methyl-114-indole-3-carboxylate (500 mg, 2.65 mmol), phenylboronic acid (384 mg, 3.17mmol), diaeetylcopper (453 mg, 3.98 mmol), triethylamine (0.44 ml, 3.98 mmol), N,N-dirnethylpyridin-4-amine (486 ml, 3.98 mmol) and 4A molecular sieve (1.02 g) in dichloromethane (15 mL) was stirred at room temperature for 12 hours. After filtration, the mixture was concentrated and purified by chromatography (silica gel, petroleum: ethyl acetate =
10:1) to afford methyl 2-methyl-I -phenyl-1H-indole-3-carboxylate as white solid (272 mg, 39%).1H NMR (400 MHz, CDC13) 6 8.19 (d, J= 8.0 Hz, 1H), 7.66¨ 7.51 (m, 3H), 7.36 (dd, J =
5.3, 3.2 Hz, 2H), 7.30 (s, 111), 7.21 ¨7.14 (m, 1H), 7.04 (d, J = 8.2 Hz, 1H), 4.00 (s, 3H), 2.62 (s, 3H).
1005381 Intermediate 4. Intermediate 4 was synthesized according to the scheme below:
Me02C
N 1 Br i,Cu20, 0s2CO3, DMF, 110 C


ii,NaH/DNIF,rt Step 1 To a solution of 2-iodopyridin-3-amine (500 mg, 2.27 mmol), cuprous oxide (32 tug, 0.23 mmol), cesium carbonate (740 mg, 2.27 mmol) in N,N-dimethylformamide (100 mL) was added methyl methacrylate (290 mg, 2.5 mmol). The reaction solution was stirred at 110 C for 12 hours. Then the reaction mixture was cooled to room temperature and added sodium hydride (60% in oil, 91 mg, 2.27 mmol) under ice bath. The resulting mixture was stirred at room temperature for half an hour. Then (1-bromoethypbenzene (418 mg, 2.27 mmol) was added.
Then the mixture was stirred at room temperature for 1 hour. After the reaction was completed, it was quenched with water (100 mL), and extracted with ethyl acetate (50 mLx 3).
The combined organic phase were washed with water (20 mLx 3) and brine (20 mL), dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (silica gel, petroleum ether/ethyl acetate = 6:1) to give methyl 2-methyl- 1-(1-phenylethyl)-1H-pyrrolo[3,2-b]pyridine-3-carboxylate (80 mg, 12 /0). LRMS (M + H-) m/z: calcd 295.14; found 295.
1005391 intermediate 5. Intermediate 5 was synthesized according to the scheme below:
CO2Me Pd(PPh3)4 CO2Me \ Br + 110 B(OH)2 Cs2CO3, dioxane, H20 .=.
Step 1 \
Methyl 2-bromo-1H-indole-3-carboxylate (200 mg, 0.79 mmol), phenylboronic acid (122 mg, 1 mmol), tetralcis(triphenylphosphine)palladium (231 mg, 0.2 mmol) and cesiumcarbonate (652 mg, 2 mmol) in dioxane/water (20 in.L/ 5 mL) were stirred at room temperature
15 hours.
Filterated the solid and the solvent was concentrated in vacuum. The resulted residue was purified by silica gel column (ethyl acetate: petroleum ether=1:2) to obtain methyl 2-pheny1-1H-indole-3-carboxylate as a pale yellow solid (160 mg, 80%) LRMS (M + Fr) m/z:
calcd 251.1 found 251.
[005401 intermediate 6. Intermediate 6 was synthesized according to the scheme below:

CO2Me CO2Me Br Cs2CO3 or Nall Br DMF \ Br 7ZnBr \ ___________________________________ 111, N 1110 Step 1 MeO,Cc Pci(PP113)4 Cs2CO3, THF, reflux (--_________ ). N
Step 2 1 =

1005411 Step 1: methyl 2-bromo-1-(1-phenylethyl)-1H4ndole-3-carboxylate. To a solution of methyl 2-bromo-1H-indole-3-carboxylate (1.0 g, 4.0 mmol) in dimethylformamide was added sodium hydride (0.32 g, 8.0mmo1, 60% in oil) under ice bath. The resulting mixture was stirred at room temperature for 0.5 hour. Then (1-bromoethyl)benzene (1.1 g, 6.0 mmol) was added in portion slowly. Then the mixture was stirred at room temperature for 1 hour.
After the reaction, it was diluted with water (100 mL), and the product was extracted with ethyl acetate (100 m1,), dried over anhydrous sodium sulfate, concentrated and purified by column Chromatography (silica gel: ethyl acetate: petroleum ether- 1:8) to give the pure product methyl 2-bromo-1-(1-phenylethyl) -1H-indole- 3-carboxylate (1.1 g, 75 /0) as a yellow oil.
1005421 Step 2: methyl 2-cyclopropy1-1-(1-phenylethyl)-111-indole-3-carboxylate. To a solution of methyl 2-bromo-1-(1-phenylethyl)-111-indole-3-carboxylate (0.30 g, 0.84 mmol) and tetrakis(triphenylphosphinc) palladium(0) (0.09 g, 0.08 mmol) in tctrahydrofuran (20 inL) was added cyclopropylzinc(II) bromidc(6.0 inL, 0.5 mol / L in tctrahydrofuran).
The reaction mixture was stirred at reflux for 12 hours. After the reaction, it was cooled to room temperature and diluted with water (100 inL). The product was extracted with ethyl acetate(100 inL), dried over anhydrous sodium sulfate, concentrated and purified by column chromatography (silica gel: ethyl acetate: petroleum ether= 1: 10) to give the crude product methyl 2-cyclopropy1-1-(1-phenylethyl)-1H-indole-3-carboxylate (0.22 g, 82%) as a yellow oil.
[005431 Intermediate 7. Intermediate 7 was synthesized according to the scheme below:

CO2Me Pa(PPh3)4 CO2Me \ Br 4" 9 Cs2CO3, aioxane, H201, Pd/C, H2, Me0H
________________________________________________________ IP
Step 1 ="'" N Step 2 CO2Me N\
[00544j Step 1: methyl 2-vinyl-I II-indole-3-carboxylate. Methyl 2-bromo-1H-indole-3-carboxylate (100 mg, 0.19 mmol), 4,4,5,5-tetramethy1-2-vinyl-1,3,2-dioxaborolane (154 mg, 1 mmol), tetrakis(triphenylphosphine)palladium (116 mg, 0.1 mmol) and cesiumcarbonate (326 mg, 1 mmol) in dioxandwater (20 mL/ 5 mL) were stirred at room temperature for 15 hours.
Filterated the solid and the solvent was concentrated in vacuum. The resulted residue was purified by silica gel column (ethyl acetate/petroleum ether = 1/2) to give methyl 2-viny1-1H-indole-3-carboxylate as a pale yellow solid (60 mg, 76%) LRMS (M + fr) m/z:
calcd 201.1 found 201.
1005451 Step 2: methyl 2-ethyl-111-indole-3-carboxylate. Methyl 2-viny1-1H-indole-3-carboxylate (80 mg, 0.4 mmol) and 10% palladium on charcoal (100 mg) in methanol (30 mL) were stirred under hydrogen 0.2 MPa at room temperature for 15 hours. The mixture was filterated the solid and concentrated in vacuum to obtain methyl 2-ethyl-1H-indole-3-carboxylate as a white solid (79 mg, 99%) LRMS (M + ) m/z: calcd 203.1 found 203.
(005461 Intermediate 8. Intermediate 8 was synthesized according to the scheme below:
s, ,o 11:Hc -"Her 14211.160 WM, methanol OH -a P4 0 sak _______________________________________________________________ 39.
EDC, HOBt T(0-Pr)4, THF
TEA, CHICI2 S, CI 1 MN' HCIMIxoane, CH2Cl2 CI' -14 N HCI>ONH ...
m DEKA. Et0H PdC12, LiCt, K2CO3 Cl .. N
KiN
DMF, 120 C
sak Intermediate 8 Step 1: N-niethoxy-N-methyltetrahydro-2H-pyran-4-carboxamide. To a suspension of tetrahydro-2H-pyran-4-carboxylic acid (9.0 g, 69.2 mmol) in CH2Cl2 (300 mL) was added N,0-dimethylhydroxylamine hydrochloride (8.05 g, 83.0 mmol), 1-(3-dimethylaminopropy1)-3-ethyl carbodiimide (20.0 g, 103.8 mmol) and triethylamine (17.5 g, 173.0 mmol) at 29 et under N2.
The reaction was stirred at 29 C for 24 hrs. Then the mixture was filtered and the the filtrate was washed with 1M ICI and saturated aqueous 1 aHC:03. The organic layer was concentrated in vacuo to give N-methoxy-N-methyltetrahydro-2H-pyran-4-carboxamide (7.1 g, 59.7%) as a yellow oil. Ill NMR (CDC13, 400 M Hz) 8 4.02-3.98 (m, 2H), 3.69 (s, 3H), 3.47-3.41 (m, 2H), 3.17 (s, 3H), 2.90 (t, J= 11.6 Hz, 3H), 1.90-1.82 (m, 2H), 1.81-1.62 (m, 2H).
[005471 Step 2: (1-(tetrahydro-2H-pyran-4-yl)ethanone. To a solution of N-methoxy-N-methyltetrahydro-2H-pyran-4-carboxamide (3.5 g, 20.2 mmol) in tetrahydrofiwan (30 inL) was added methylmagnesium bromide (12 mL, 36.4 mmol) at -65 C under N2. The reaction was stirred at -65-15 C for 3 hrs. Then the mixture was quenched by water, extracted with ethyl acetate. The organic layer was washed with 1M HC1, saturated aqueous NaHCO3 and concentrated in vacuo to give 1-(tetrahydro-2H-pyran-4-yl)ethanone (1.9 g, 73.6%) as a yellow oi1.1H NMR (CDC13, 400 M Hz) 6 4.01-3.97 (m, 2H), 3.45-3.39 (m, 3H), 2.57-2.49 (m, 1H), 1.80-1.71 (m, 2H), 1.70-1.62 (m, 2H).
[005481 Step 3: aR,E)-2-methyll-N-(l-(tetrahydro-2H-pyran-4-y1)ethylidene)propane -2-sulfinamide). To a solution of 1-(tetrahydro-2H-pyran-4-yl)ethanone (1.9 g, 14.8mm01) in tetrahydroftwan (30 inL) was added (R)-2-methylpropane-2-sulfinamide (2.15 g, 17.8mmo1) and tetraethoxytitanium (5.06 g, 22.2mmo1) at 25 C under N2. The reaction was refiuxed for 7 hrs.
Then the mixture was quenched by water, extracted with ethyl acetate and filtered. The crude product was purified by column chromatography on silica gel eluted with dichloromethane:
methanol= 200:1 to give (R,E)-2-methyl-N-(1-(tetrahydro-2H-pyran-4-yi)ethylidene)propane-2-sulfinamide (1.5 g, 44.1%) as a yellow oil.
1005491 Step 4: ((R)-2-methyl-N-(1-(tetrahydro-211:11-pyran-4-yl)ethyl)Propane--sulfmamide). To a solution of (R,E)-2-methyl-N-(1-(tetrahydro-2H-pyran-4-yDethylidene)propane-2-sulfinamide (1.25 g, 5.4 mmol) in tetrahydrofuran (6 mL) was added methanol (692 mg, 21.6 mmol) and NaBH4 (821 mg, 21.6 mmol) at -65 C under N2.
The reaction was stirred at -65-0 C for 2 h. Then the reaction was quenched by water and extracted with ethyl acetate. The organic layer was washed with saturated aqueous NaHCO3 and concentrated in vacuo to give crude (R)-2-methyl-N-(1-(tetrahydro-2H-pyran-4-ypethyl)Propane-2-sulfinamide (1.2 g, 52.1%) as a yellow oil.

[005501 Step 5: ((S)-1-(tetrahydro-2H-pyran-4-yi)ethanamine). To a solution of (R)-2-methyl-N-(1-(tetrahydro-2H-pyran-4-ypethyl)Propane-2-sulfinamide (1.2 g) in CH2Cl2 (4 mL) was added HC1-dioxane (4 mL) at 25 C. The reaction was stirred for 2 hrs.
Then the mixture was concentrated in vacuo, diluted with water and washed with ethyl acetate to give crude (S)-1-(tetrahydro-2H-pyran-4-yl)ethanamine (1.0 g) as a yellow oil.
[005511 Step 5: ((S)-5-bromo-2-chloro-N-(1-(tetrahydro-2H-pyran-4-ypethyl)Pyrimidin -4-amine). To a solution of (S)-1-(tetrahydro-2H-pyran-4-yl)ethanamine (1.0 g, 7.75 nunol) in ethanol (15 mL) was added N,N-diisopropylethylarnine (3.0 g, 23.3 mmol) and 5-bromo-2,4-ichloropyrimidine (2.0 g, 8.53 mmol) at 25 C under N2. The reaction was stirred at 25 C for 18 hrs. The mixture was diluted with ethyl acetate and washed with water. The crude product was purified by column chromatography on silica gel eluted with petroleum ether:
ethyl acetate =
20:1 to give (S)-5-bromo-2-chloro-N-(1-(tetrahydro-2H-pyran-4-yl)ethyl)Pyrimidin-4-amine (1.5 g, 60.7%) as a yellow oil.
[005521 Step 6: ((S)-ethyl 2-chloro-6-methyl-7-(1-(tetrahydro-2H-pyran-4-yi)ethyl)-7H-pyrrolo(2,3-d1pyrimidine-5-carboxylate), Intermediate 8. To a solution of (S)-5-bromo-2-chloro-N-(1-(tetrahydro-2H-pyran-4-yl)ethyl)Pyrimidin -4-amine (1.5 g, 4.7 mmol) in D11417 (20 mL) was added ethyl but-2-ynoate (1.05 g, 9.4 mmol), lithium chloride (296 mg, 7.05 mmol), palladium(II) acetate (105 mg, 0.47 mmol) and potassium carbonate (1.95 g, 14.1 mmol) at 25 C
under N2. The reaction was stirred at 120 C for 3 hrs. The mixture was filtered and the crude product was purified by column chromatography on silica gel eluted with petroleum ether: ethyl acetate = 30:1 to give (S)-ethyl 2-chloro-6-methy1-7-(1-(tetrahydro-2H-pyran-4-yl)ethyl)-7H-pyrrolo[2,3-d]pylimidine-5-carboxylate (270 mg, 16.4%) as a yellow oil.
[005531 Intermediate 9. Intermediate 9 was synthesized according to the scheme below:

0 OH i H
HATLI/TENDCM
MetAgBrITHF/-60 C- reductive amination PMB'N6 A- H2,Pci/C

N Stepl Step 2 I N N N
N
Boo Bico Bloc 1 Boo Boo OEt 0,....0Et Br N cr-Clei 6 I.4 N H-A50PsiVP&CIMIDHIrt ___.... ..), Pd(Ac0)26_10V1(2003P1 t DPENEt0H/rt CI N N
H.'-'0NBoc Dtt4F/120 C/MW Step 7 Step 5 Step 6 Bocte- BoolOiss Intermediate 9 [005541 Step 1: tert-butyl 4-(methoxy(methytcarbamoyDpiperidine-1-carboxylate.
To a solution of 1-(tert-butoxycarbonyl)piperidine-4-carboxylic acid (5.0 g, 21.8mmo1) in dichloromethane (80mL) were added N,0-dimethylhydroxylamine hydrochloride (2.55 g, 26.1rnmol), 2-(3H- [1,2,3]triazolo [4,5-b]pyridin-3-y1)-1,1,3,3-tetramethylisouroni um hexafluorophosphate(Y) (9.95 g, 26.1mmol), N-ethyl-N-isopropylpropan-2-amine(8.46g, 65.42mmo1) and stirred at 30.0 for 16 hours. The reaction was quenched by adding water (100m1) and extracted with ethyl acetate (200 ml*3). The combined organic layers was washed with brine, dried over sodium sulfate and concentrated under vacuum to give tcrt-butyl 4-(methoxy(methyl)carbamoyl)piperidine-l-carboxylate.(5.7 g, yield 97.6%). LRMS
(M + H ) m/z: calcd 272.17; found 273.17.
[005551 Step 2: tert-butyl 4-acetylpiperidine-1-carboxylate. To a solution of tert-butyl 4-(methoxy(methyl)carbamoyl)piperidine-1-carboxylate (4.0 g, 14.69mmol) in tetrahydrofuran (50 mL) was added methylmagnesium bromide solution (5.25 g, 44.06 mmol, 3M) at -70 C over 0.5 hour. The resultant mixture was stirred at room temperature for 16 hours. The reaction solution was adjusted pH to 6.0 by LOM hydrochloride solution. The aqueous phase was extracted with ethyl acetate(100m1*3). The combined organic layers was washed with brine, dried over sodium sulfate and concentrated under vacuum to give tert-butyl 4-acetylpiperidine-l-carboxylate as a yellow liquid (2.6 g, yield 77.8 %). LRMS 04 ti+) nth: calcd 227.15; found 228.
[005561 Step 3: tert-butyl 4-(1-aminoethyl)piperidine-l-carboxylate. To a solution of tert-butyl 4-acetylpiperidine-1-carboxylate (10g, 44.05mmo1) in tetrahydrofuran (100m1) and methanol (100m1) were added ammonium acetate (20g, 264.3nunol) and sodium cyanotrihydroborate(20g, 264.3mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuum. The crude product was added wateg100m1), and extracted with ethyl acetate (50mL x3) . The organic layer was dried over sodium sulfate and concentrated to give tert-butyl 4-(1-aminoethyppiperidine-l-carboxylate (6g, yield: 60%) as a colorless liquid. LRMS (M-I-H4) m/z: calcd 228.15; found 229.
[005571 Step 4: tert-butyl 4-(1.-((5-bromo-2-ehloropyrimidin-4-yDamino)ethyppiperidine-1-carboxylate. To the solution of tert-butyl 4-(1-aminoethyl)piperidine- 1 -carboxylate (5 g, 21.9 mmol) in ethanol (50 ml) were added 5-bromo-2,4-dichloropyrimidine (5.97 g, 26.3 mmol), and N-ethyl-N-isopropylpropan-2-amine (8.48g ,65.7mmo1). The resulting mixture was stirred at room temperature overnight.
The reaction mixture was concentrated in vacuum, and the crude product was added to water (100m1). The aqueous layer was extracted with ethyl acetate (50rnL x4) and organic layer was dried over sodium sulfate and concentrated to give crude product tert-butyl 4-0-((5-bromo-chloropyrimidin-4-yl)amino)ethyl)piperidine-l-carboxylate (7 g, yield:
97.2%)as a yellow oil.
LRMS (M-FH+) ,n/z: calcd 418; found 419.
[00558] Step 5: ethyl 74141 -(tert-butoxycarbonyl)piperidin-4-yl)ethyl)-2-chloro-6-methyl-7H-pyrrolol2,.3-d 1pyrimidine-5-ea rboxylate. Into the vial were added tert-butyl 441-((5-bromo-2-chloropyrimidin-4-yl)amino)ethyl)piperidine-l-carboxylate (400mg, 0.95mmo1) and ethyl but-2-ynoate ( 128mg, 1.14mmol ) , palladium(H) diacetate (42mg, 0.19mmol), lithium chloride (51.8mg, 1.23mmol), carbonic acid (393mg,2.85mmo1) in N, N-dimethyl formamide. The mixture was degassed for 10min and refilled with nitrogen, and irradiated in the microwave on a Biotage smith synthesizer at 120 C for 40min.The reaction mixture was cooled down and added to water (30m1), and extracted with ethyl acetate(10m1x3). The organic layer was dried over sodium sulfate and concentrated. The crude product was purified by column chromatography on silica gel (petroleum/ethyl acetate: 20:1 to 10:1) to give compound ethyl 7-(1-(1-(tert-butoxycarbonyl)piperidin-4-ypethyl)-2-chloro-6-methyl-7H-pyrrolo [2,3-d]pyrimidine-5-carboxylate(75mg, yield :17.4%) as a yellow oil. LRMS (M+11.4) m/z: calcd 450.2; found 451.

1005591 Step 6: tethyl 7-(1-(1-(tert-butoxycarbonyl)piperidin-4-Aethyl)-6-methyl-7H-pyrrolo12,3-dipyrimidine-5-carboxylate, Intermediate 9. A mixture of compound ethyl 7-(1-(1-(tert-butoxycarbonyl)piperidin-4-yl)ethyl)-2-chloro-6-methyl-7H-pyrrolo [2,3-d] pyrimidine-5-carboxylate (75 mg, 0.16 mmol) and Pd/C(10mg) in methanol (10mL) was stirred under 50psi of hydrogen at room temperature overnight. The reaction mixture was filtered and the filtrate was concentrated to give product ethyl 7-(1-(1-(tert-butoxycarbonyl)piperidin-4-ypethyl)-6-methyl-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylate (50mg,yield :72%). LRMS (M F14.) m/z: calcd 450.2; found 451.
1005601 Intermediate 10. Intermediate 10 was synthesized according to the scheme below:
Ho ;4*F F _NH HO MervIgBr H2N
Na AcCINH4 )----0(FF
__________________________________________ HATU DIEA BH3CN
Intermediate 10 [005611 Step 1: 4, 4-difluoro ¨N-methyl cyclohexanecarboxamide. To a solution of 4 , 4-difluorocyclohexanecarboxylic acid (5 g, 30.49 mmol) in DCM (40 mL) were added DIPEA
(11.8 g, 91.46 mmol) and HATU (17.38 g,45.73 mmol). The mixture was stirred for 0.5 hr. The NO-dimethylhydroxylamine hydrochloride (3.56 g , 36.58 rmnol) was added to the mixture. The mixture was stirred at 28 C for 12 hr. Water (20mL) was added and the mixture was extracted by CH2Cl2 (30 mL*3). The organic layer was washed with brine and dried over sodium sulfate. The organic layer was concentrated to give the crude product, the crude product was purified by column chromatography on silica gel eluted with ( petroleum ether / ethyl acetate 10:1¨*2:1) to gave 4, 4-difluoro --N-methyl cyclohexanecarboxamide (4.6 g, 73.36 %) for next step.
1005621 Step 2: 1 -(4,4- difluorocyclohexyl) ethanone. To a solution of 4,4-difiuoro -N-methyl cyclohexanecarboxamide (3 g, 14.5 mmol) in THF (20 mL) were added CH3MgBr (5.19 g, 43.5mmo1 ) in -78 C. The mixture was stirred at -78 C for 5 hr. Water (10mL) was added and the mixture was concentrated and extracted by CH2C12 (40 mL*3). The organic layer was washed with brine and dried over sodium sulfate. The organic layer was concentrated to give 1 -(4,4- difluorocyclohexyl) ethanone (1.5 g, crude) for next step.
1005631 Step 3: 1 -(4,4- difluorocyclohexyl) ethanamine, Intermediate 10. To a solution of 1-(4, 4- difluorocyclohexyl) ethanone (1 g, 6.17 mmol) in THF (5mL) was added a solution of ammonium acetate (9.51 g, 123.4 mmol) and NaBH3C'N (3.82 g, 61.7 mmol) in Me0H. The mixture was stirred for 48 hr. The reaction mixture was quenched with aqueous Na1-1CO3 and extracted with tert-Butyl-methyl ether (20 mL*3) .The combined organic layers were washed with IN HCI ,and the organic and aqueous layer were separated .The aqueous layer was brought to above pH 8 with 2N NaOH and extrated with ethyl acetate(20 mL*3) . The organic layer was washed with brine, dried over sodium sulfate. The organic layer was concentrated to give 1 -(4,4-difluorocyclohexyl) ethanarnine (545 mg, crude ) for next step.
[005641 Intermediate 11. Intermediate 11 was synthesized according to the scheme below:
f¨\
Boc¨N p H a, Boc¨N"'0--\ MeMgBr 0 Boc¨N 0 TFA/DC*I
0 b 0 CbzCI Cbz¨N 0 HN 0 Cbz¨N 0 AcONH4 FIN
NaBH3(CN) step 3 Cf 0 2 days HõN
Intermediate 11 [005651 Step 1: tert-butyl 2-(metboxy(methyl)carbamoyi)morpholine-4-carboxylate. To a solution of 4-(tert-butoxycarbonyl)morpholine-2-carboxylic acid (10 g, 43 mmol) in DCM (60 mL) was added FIATU (19 g, 50 mmol) in portions, The mixture was stirred at room temperature for 0.5h. Then DIPEA (12.9 g, 100 mmol) and N, O-dimethylhydroxylamine hydrochloride (5.0 g, 51.5 mmol) were added in the mixture. The mixture was stirred at room temperature overnight. Water (40 mL) was added and the mixture was extracted by DCM (100 mL*3). The organic layer was washed with brine (5 mL) and dried over sodium sulfate.
Concentration and purification by column chromatography on silica gel (eluted:
petroleum ether/ethyl acetate = 20/1-10/1) to give tert-butyl 2-(methoxy(methyl)carbamoyl)morpholine-4-carboxylate (10 g, 84%).
1005661 Step 2: tert-butyl 2-acetylmorpholine-4-carboxylate. To a solution of tert-butyl 2-(methoxy(methyl)carbamoyl)morpholine-4-carboxylate (11 g, 4.0 mmol) in THF (40 mL) was added CH3MgBr (4 mL, 12 mmol) at -78 C. The mixture was stirred at -78 C for 3 hr. Water (20 mL) was added and the mixture was extracted by ethyl acetate (80 mL*3).
The organic layer was washed with brine and dried over sodium sulfate. The crude product was concentrated and purified by column chromatography on silica gel (eluted: petroleum ether/ethyl acetate = 10/1) to give tert-butyl 2-acetylmorpholine-4-carboxylate (8 g, 86%).
[005671 Step 3: 1-(morpholin-2-yl)ethanone. To a solution of tert-butyl 2-acetylmorpholine-4-carboxylate (6.9 g, 30.0 mmol) in DCM (80 mL) was added TFA
(20 mL).
The mixture was stirred at room temperature for 4 hr. The reaction mixture was concentrated to give 1-(morpholin-2-yl)ethanone (3.9 g, crude ) for next step.
[005681 Step 4: benzyl 2-acetylmorpholine-4-carboxylate. To a solution of 1-(morpholin-2-yl)ethanone (3.9 g, 30.0 mmol) in DCM (60 mL) was added triethylamine (1.0 g, 10 mmol) to adjust pH 9. Then benzyl carbonochloridate (5.6 g, 33.0 mmol) and triethylamine (3.0 g, 30.0 mmol) were added and the mixture was stirred at room temperature for 16 hr.
The reaction solution was added 1120 (20 mL) and extracted by DCM (100 mL*3). The organic layer was concentrated and purified by column chromatography on silica gel (eluted:
petroleum ether/ethyl acetate = 20/1-15/1-10/1) to give benzyl 2-acetylmorpholine-4-carboxylate (5.0 g, 63 %).
[005691 Step 5: benzyl 2-(1-aminoethyl)morpholine-4-carboxylate, Intermediate 11. To a solution of benzyl 2-acetylmorpholine-4-carboxylate (5.0 g, 19 mmol) in THF/MeOFT (1/5, 100 mi.) were added Ac0NH4 (14.6 g, 190 mmol) and NaBl-T3CN (11.8 g, 190 mmol).
The mixture was stirred at room temperature for 50 hr. The mixture was concentrated to give the crude. Water (20 mL) was added and the mixture was extracted by ethyl acetate (60 mL*3). IN
HC1.was added into the combined organic layers. The aqueous layer was concentrated, brought to above pH >8 with 2N NaOH and extracted by ethyl acetate (60 mL*3). The organic layer was washed with brine, dried over sodium sulfate. The mixture was concentrated to give benzyl 2-( 1 -aminoethyl)morpholine-4-carboxylate (2.0 g, 40 %).
(00570) Example 75. measurements for Inhibitors using EZH2.
[005711 EZI12 Assay: Assays were carried out by mixing rPRC2 together with biotinylated oligonucleosome substrates in the presence of the radio-labeled enzyme co-factor, S-adenosyl-L-methionine (3H SAM) (Perkin Elmer) and monitoring the enzymatically mediated transfer of tritiated methyl groups from 3H SAM to histone lysine residues. The amount of resulting vitiated methyl histone product was measured by first capturing the biotinylated oligonucleosomes in streptavidin (SAV) coated FlashPlates (Perkin Elmer), followed by a wash step to remove un-reacted 3H SAM, and then counting on a TopCount NXT 384 well plate scintillation counter (Perkin Elmer). The final assay conditions for EZH2 were as follows: 50 mM Tris Buffer pH 8.5, 1 niM DTT, 691.IM Brij-35 detergent, 5.0 mM MgC12, 0.1 mg/mL BSA, 0.2 1.t.M 3H SAM, 0.2 p.M biotinylated oligonucleosomes, 3.6 p.M H3K27me3 peptide and 2 nM
EZH2.
1005721 Compound IC50 measurements were obtained as follows: Compounds were first dissolved in 100% DMSO as 10 mM stock solutions. Ten point dose response curves were generated by dispensing varying amounts of the 10 intM compound solution in 10 wells of the 384 well plate (Echo; Labcyte), pure DMSO was then used to backfill the wells to insure all wells have the same amount of DMSO. A 12.5 p1 volume of the HMT enzyme, H3K27me3 peptide and oligonucleosome substrate in assay buffer was added to each well of the assay plate using a Multidrop Combi (ThermoFisher). Compounds were pre-incubated with the enzyme for 20 min, followed by initiation of the methyltransferase reaction by addition of 12.5 111_, of 3H
SAM in assay buffer (final volume = 25 ttL). The final concentrations of compounds ranged from a top default concentration of 80 glµil down to 0.16 1.tM in ten 2-fold dilution steps.
Reactions were carried out for 60 minutes and quenched with 20 pi, per well of 1.96 mM SAIL
50 mM Tris pH 8.5, 200 mM EDTA. Stopped reactions were transferred to SAV
coated Flashplates (Perkin Elmer), incubated for 120 min, washed with a plate washer, and then read on the TopCount NXT (1.0 min/well) to measure the amount of methyl histone product formed during the reaction. The amount of methyl histone product was compared with the amount of product formed in the 0% and 100% inhibition control wells allowing the calculation of %
Inhibition in the presence of the individual compounds at various concentrations. IC50's were computed using a 4 parameter fit non-linear curve fitting software package (XLEET, part of the database package, ActivityBase (IDBS)) where the four parameters were IC50, Hill slope, pre-transitional baseline (0% INH), and post-transitional baseline (100% INH);
with the latter two paratneters being fixed to zero and 100%, respectively, by default.
1005731 Assay for Y641N EZH2 was performed as above using reconstituted H3K27Me2 oligonucleosomes as substrate.
1005741 Table 2 shows the activity of selected compounds of this invention in the EZH2 and Y641N EZH2 activity inhibition assay. IC50 values are reported as follows: "A"
indicates an IC50 value of less than 100 nM; "B" indicates an IC50 value of 100 nM to 1 p.M; "C" indicates an IC50 value of greater than 1 IAM and less than 10 p.M for each enzyme; "D"
indicates an IC50 value of greater than 10 gM for each enzyme; and "*(X AM)" indicates that no inhibition was observed at the highest concentration (i.e., X gM) of compound tested.
Table 2. 1050 Values for Compounds of Formula I against EZH2 and Y641N EZH2 Mutant Enzymes.
Compound EZH2 IC,,r, 1 Y641.N Compound No. ' 1 EZH2 IC50 No. EZ112 1050 100 C i D 140 101 D D ___________ 141 B C

105 B . C 145 A B

09*(1014M) *(10gM) 149 B C
iiii- -*(Ioj.A) *(I OgM) 150 B B
111 A A ___________ 164 _______ A B
112 *(10g4) *(10gM) 165 .A B
113 B C ___________ 166 ______ A B
114 .A A 167 B B

117 A B , 170 A B
118 C *(1.0gM) 171 A A

121 *(0.511,M) *(10gM) 173 A B

124 176 A B , ¨ _ 131 , *(1.01tM) *(1.0gM) 181 A A
132 A B -------- ------- ----- * .
182 (10gM) *(0.504) 133 A . B 183 A A -----, -----137 A A 187 A B -----, 138 B C , 188 , B C
139 B C 189 B _ C

Compound E IC Y641.N Compound Y641N

No. = EZH2 IC5o No. , EZH2 IC5 EZH2 .IC5o 192 .A B 235 B C
193 B C 236 *(0.5p,M) , *(101.1M) 195 B *(0.501) 238 B C

- , 202 B C 245 A , B

204 A B 247 A ________ A
¨ .....

206 13 C 249 B , C
207 A. B 250 A B
208 B B 251 B B ...
209 C *(1.0p.N1) 252 A B
210 A B 253 A , B

_212 A B 255 A C
ET 7-A- ii- 256 A B
214 B C 257 B , c 217 B B __________ 260 B C
_ ....._ 218 A A 261 A , A

1/5 A A 268 A B i , 227 A _______ A 271 C C
_ //8 B B 272 C *(10p,N4) 229 C D 273 _ A A

231 , A B 275 A , A.

233 B C 277 A i A

Compound Y641.N Compound Y641N
EZH2 ICso EZH2 ICso No. EZH2 iCso No. , EZH2 .1C5o 279 .A B 323 A B

281 A A 326 A A.

286 A ______ B 332 A A
287 El . C 333 A B

290 A B 335 A A.

29/ A B __________ 337 A A

299 A A 344 A A.
300 A A ......._345 A A ....._ 303 A. B 348 A A.

A
305 A B ____________ 350 A
.... _ _ 309 El C 354 A A

311 A A 356 A A.

315 A B __________ 360 A A ..

317 A A 362 _ A A

319 A _______ A 364 . A B

321 ---------- A A ___________ 366 A 1 A ., Compound Y641.N Compound Y641N

No. EZH2 IC50 No. EZH2 368 .A A 376 A A

371 A A 445 *(10u4) *(10uM) 372 B *(0.5uM) 446 *(10uM) *(10uM) [005751 EXAMPLE 76. EC MEASUREMENTS FOR INHIBITORS IN HELA
CELL ASSAYS.
[005761 H3K27me3 MSD Hela Assay. Trypsinized HeLa cells were counted and diluted in 10% DMEM (Life Technologies, Cat. # 10569) to 5000 cells/75 pt. Seventy-five pi, of cells were place in each well of a 96-well flat-bottomed plate and incubated at 37 C
for 4 hours.
Twenty-five 'IL of test compound (at various concentrations) was added to the cells and incubation continued at 37 C for 96 hours. Media was then removed and the cells rinsed once with ice cold PBS. Forty uL of ice-cold MSD Butler AT (10 mM HEPES, pH 7.9, 5 mM
MgCl2, 0.25M sucrose, Benzonase (1:10000), 1% Triton X-100 supplemented with fresh lx Protease Inhibitor cocktail and 1mM 4-(2-Aminoethyl)benzenesulfonyl fluoride hydrochloride (AEBSF)) was added to each well and the plat; placed on ice for 30 minutes.
Ten ILL of 5M
NaCi was then added to each well and incubation on ice continued for another 15 minutes. The material in each well was suspended pipetting up and down and then transferred to a new 96 well plate. The emptied wells were rinsed with 150uL ice-cold 20mM Tris pH 7.5, 1mM
EDTA, 1mM EGTA, supplemented with fresh lx Protease Inhibitor cocktail and 1mM AEBSF
("NO
salt NO detergent buffer) and transferred to the respective wells in the new plate. Three hundred p.I., of NO Salt NO detergent buffer was then added to each well of lysates and the plates frozen at -80 C.
1005771 On the same day, an appropriate number of MSD standard bind 96-well plates were coated with 304/well of total H3 capture antibody (Millipore, Cat # MAB3422) at 1 ug/mL
concentration in PBS. The antibody solution was evenly distributed first by tapping gently on the sides of the plates and then by shaking the plates for a few minutes at 1000 rpm. Antibody coated. plates were stored at 4 C overnight.

[005781 The next day the lysates are thawed to RT. The antibody coated MSD
plates are washed 3X with TBS-T (Tris-buffered saline (Fisher Scientific, Cat #BP2471-1) + 0.2% Tween-20). One-hundred fifty AL of 5% Blocker A in TBS-T is added to each well. The wells are covered and shaken on a shaker at RT for one hour. The Blocker A step is repeated a second time. After removing the blocker, 25 AL of cell lysate is transferred into each antibody coated well. The plates are shaken for 2 hours at RT, the lysate removed and the plates again washed with Blocker A in TBS-T. Twenty-five AL of appropriate freshly prepared antibody mix (including both primary and secondary antibodies) is added to each well and the plates shaken for 1 hour at RT. The antibody mix used was one (or both) of those indicated in the table below:
Ab Concentration Primary Anti-rabbit detection 1% blocker A
(1.1g/1nL) Ab (AL) Ab (AL) (4) H3K27me3 33 37.88 5.00 5000 H3 12 52.08 5.00 5000 Both H3 antibodies were obtained from Cell Signalling (Cat #s 4499 and 9733).
The goat anti-rabbit antibody was obtained from Meso-Scale Discovery (Cat #R32AB-1).
1005791 The antibody mix was then removed and the wells washed with Blocker A.
One hundred-fifty Al of freshly prepared 1 X MSD Read Buffer (Meso-Scale Discovery; Cat #R927C-2) was then added to each well and the plates read on a MSD Sector 2400 Plate Reader.
100580] Data was analyzed using Assay Assistant (Constellation Pharmaceuticals In-house product) and Activity Base (IDBS Ltd, Surrey, UK) template. Data files were imported to Assay Assistant and assay conditions were specified. A unique Analysis ID was created and the data files exported to Activity Base. An analysis template was created on Activity Base to measure dose-dependent inhibition of H3K27me3 mark and cell viability respectively.
Readout of DMSO
wells were used to normalize the data. Resulting curves were fitted using Activity base software Model 205 (IDBS Ltd, Surrey, UK). The data was checked for quality, validated and integrated in excel format using SARview (IDBS Ltd, Surrey, UK).
[005811 H3K27me3 Alpha Hela Assay (AlphaLISA). Ten different doses of each test compound (in a series of 3-fold dilutions) were plated in duplicate 34-well tissue culture treated plates (Catalog # 781080; Greiner Bio One; Monroe, North Carolina). Hela cells grown in culture were trypsinized and counted using a Countess cell counter (Catalog #
C10281; Life Technologies, Grand Island, NY). Cell were diluted to 67,000 cells per mi.
in10% DMEM

(Catalog ft 10569-010 Life Technologies, Grand Island, NY) and 154 (1,000 cells) were plated into each well using the Biotek MicroFlomi Select Dispenser (BioTek Instruments, Inc.
Vermont, USA),) of the 384-well plate. Plates were incubated at 37 C /5% CO2 for 72 hrs. One of the duplicate plates was processed for HeLa assay and the other for viability.
1005821 To the plate processed for AlphaLISA was added 5AL per well Cell-Histone Lysis buffer (IX) (Catalog # ALOO9F1 Perkin Elmer; Waltham, MA) and the plate was incubated at RT for 30 minutes on a plate shaker with low speed (Model# 4625-Q Thermo Scientific;
Waltham, MA). Then, 104 per well Histone Extraction buffer (catalog #
.A.L009F2; Perkin Elmer; Waltham, MA) was added and the plate further incubated at RT for 20 min on plate shaker with low speed. To each well was then added 104 per well of a 5X mix of anti-K27me3 acceptor beads plus Biotinylated anti-Histone H3 (C-ter) Antibody (diluted to 3nM final) (Catalog #AL118 Perkin Elmer; Waltham, MA). Dilution of the acceptor beads and then anti-Histone H3 was with 1X Histone Detection buffer (Catalog # ALOO9F3 Perkin Elmer; Waltham, MA) which was produced diluted from the 10X stock provided. The plate was sealed with an aluminum plate sealer and incubated at 23 C for 60 min. We then added 104 5X
solution of Streptavidin Donor beads (Catalog #6760002 Perkin Elmer; Waltham, MA) (20 g/m1., final in 1X Histone Detection Buffer), sealed the plate with Aluminum plate sealer and incubated at 23 C for 30 min. The plates were then read using an EnVision- Alpha Reader (model # 2104 Perkin Elmer; Waltham, MA).
1005831 Cell viability was assayed by adding 15 lit of Cell Titer Ole ((Catalog #07571 Promega Madison, WI) to each well with cells with media. The plates were incubated foat RT
for 15 - 20 minutes on a plate shaker at low speed. The plates were then read using an EnVision-Alpha Reader (model # 2104 Perkin Elmer; Waltham, MA).
1005841 Data from both assays was analyzed using Assay Assistant (Constellation Pharmaceuticals In-house product) and Activity Base (IDBS Ltd, Surrey, UK) template. Data files were imported to Assay Assistant and assay conditions were specified. A
unique Analysis ID was created and the data files exported to Activity Base. An analysis template was created on Activity Base to measure dose-dependent inhibition of H3K27me3 mark and cell viability respectively. Readout of DMSO wells were used to normalize the data. Resulting curves were fitted using Activity base software Model 205 (IDBS Ltd, Surrey, UK). The data was checked for quality, validated and integrated in excel format using SARview (IDBS Ltd, Surrey, UK).

[005851 Table 3 shows the activity of selected compounds of this invention in the two different HeLa cell assays described above. EC50 values are reported as follows: "A" indicates an EC50 value of less than 400 nM; "fr indicates an EC50 value of 400 nM to 2 AM; "C"
indicates an EC50 value of greater than 2 gM and less than 10 gIVI for each enzyme; "D"
indicates an EC50 value of greater than 10 AM for each enzyme; and "*(X gM)"
indicates that no inhibition was observed at the highest concentration (i.e., X gM) of compound tested.
Table 3. Ec50 Values for Selected Compounds of the Invention In Hela Cells Expressing H3k27 Mutant EZH2.
H3K27mc3 H3K27mc3 . H3K27mc3 H3K27mc3 Alpha_ .. MSD.... Alpha_ . MSD...
Compound HeLa HeLa Compound HeLa lHeLa No. (EC50) ....
(EC50) No. (EC50) (EC56 123 B , 212 B

137 B 218 A .
143 C .

144 C 11-, ,..,..:. _________ D

147 B 230 B ___ _ 166 C 238 C .
169 D ... 239 B

171 j C 241 B

172 1 C 1 .... _..... 242 C ___ .

181 B 150 C .
187 J3 1.3 253 A

_ 113K27me3 1-13K27me3 1 H3K27me3 Ii3K27me3 _Alpha_ MSD_ I _Alpha_ MSD_ Compound HeLa HeLa_ I Compound HeLa lileLa No. AEC5.9) ........... (gC5) j No. (EC50) (EC565 281 A A . 343 B
283 B 344 A . .

288 A. B 347 B
291 B i 349 C
294 A A 351 N aN
298 A A 35; B
300 A A L353 *133 __11N/I) 31() A A 356 A
-1-- ¨1 311 B -357¨ ¨A

3.14 A 359 B i i----317 ----------------- A 362 .A

¨I
319 A 364 *(3.33 1.1.M) 321 A 366 B .

330 B 370 *(3.33 JIM) 332 B 371 *(3.33 1.1.M) _ 333 1 B 373 A .

337 A 376 N aN

339 A , ....
_..... 395S________

Claims (40)

We Claim:
1.A compound having structural formula I:
or a pharmaceutically acceptable salt thereof. wherein:
Z is =C(R2)- or =N-;
each of X1 and X2 is =C(R3)-, wherein two R3 are taken together with the carbon atoms to which they are bound to form an aryl, heteroaryl, or carbocyclyl fused to the ring comprising X1, X2 and X3;
X3 is =N- or =C(R6)-;
no more than one of X1, X2, and X3 is =N- :
each R1 and R2 is independently hydrogen. halo, -OH, -CN, C1-C4 alkyl, -O-(C1-C4 alkyl), -N(R7)2, -(C0-C4 alkylene)-aryl, -(C0-C4 alkylene) -heteroaryl, -(C0-C4 alkylene)-heterocyclyl, or -(C0-C4 alkylene)-carbocyclyl; or one R1 and R2 are taken together with atoms to which they are bound to form an aryl, heteroaryl, heterocyclyl, or carbocyclyl ring;
R6 is hydrogen, halo, -CN. -(C0-C4 alkylene)-R8, -(C2-C6 alkenyl or alkynyl)-R9, -(C1-C4 alkylene)-O-R9, alkylene)-O-(C1-C4alkylene)-R8, -O-(C0-C4 alkylene)-R9, -O-(C2-C4 alkylene)-O-R8, -O-(C1-C4 alkylene)-R9, -(C0-C4 alkylene)-N(R7)2, -(C0-C4 alkylene)-C(O)-O-R9, -(C0-C4 alkylene)-O-C(O)-R9, -(C0-C4 alkylene)-C(O)-N(R7)2, -(C0-C4 alkylene)-N(R9)-C(O)-R9, -O-(C1-C4 alkylene)-C(O)-N(R7)2, -O-(C2-C4 alkylene)-N(R9)-C(O)-(R7), -(C0-C4 alkylene)-S(O)-R8, -(C0-C4 alkylene)-S(O)2-R8 or -(C0-C4 alky1ene)-S(O)2-N(R7)2; or R8 is Q. -S(O)2-Q, -C(O)¨Q, or -CH(R4)(R5);
Q is aryl, heteroaryl, heterocyclyl, or carbocyclyl;

R4 is C2-C6 alkyl, -CH2-O-(C1-C4 alkyl) or -(C0-C6 alkylene)-Q, wherein one or two methylene units in the alkyl or alkylene portion of R4 are optionally and independently replaced by -O-, -S -, -S(=O) -S(=O)2-, or -N(R10)-;
R5 is hydrogen, -(C0-C6 alkylene)-Q, or C1-C6 alkyl, wherein one or two methylene units in R5 are optionally and independently replaced by -O-, -S-, -S(=O) -S(=O)2-, or -NR10-;
each R7 is independently -(C0-C4 alkylene)-R9, -(C0-C4 alkylene)-O-R9, -S(O)2-R8, -C(=O)-R8, -C(=O)-N(R9)2, -(C1-C4 alkylene)-O-C(=O)-R8 or -(C0-C4 alkylene)-C(=O)-O-R9;
or two R7 are taken together with the nitrogen atom to which they are commonly bound to form an optionally substituted heterocyclyl or heteroaryl ring;
R8 is C1-C4 alkyl, aryl, heteroaryl, carbocyclyl or heterocyclyl;
R9 is hydrogen or R8;
R10 is hydrogen, C1-C4 alkyl, -S(=O)2-R9, -C(=O)-R8, -C(=O)-N(R9)(R12); or -C(=O)-O-R11;
R11 is unsubstituted C1-C4 alkyl or C1-C4 haloalkyl;
R12 is hydrogen, unsubstituted C1-C4 alkyl or C1-C4 haloalkyl;
wherein unless otherwise designated any alkyl, alkylene, alkenyl, alkynyl, aryl, heteroaryl, heterocyclyl or carbocyclyl portion of the compound is optionally substituted;
and wherein:
when each R1 is methyl, Z is =C(H)-, X3 is =N-, and R x is phenyl or 4-fluorophenyl; then the R3 of X1 and the R3 of X2 are not taken together to form unsubstituted C5-C7 cycloalkyl fused to the ring comprising X1, X2 and X3.
2. The compound of claim 1, wherein one R1 is -CH3; the other R1 is -C1-C2 alkyl optionally substituted with one or more fluoro or -O-(C1-C2 alkyl) optionally substituted with one or more fluoro.
3. The compound of claim 2 represented by the formula:
wherein:
R1a is -OCH3, -CH3, -OCHF2, or -CH2CH3;

R1b is -CH3; and Z is =CH-.
4. The compound of claim 3, wherein two R3 are taken together with the carbon atoms to which they are bound to form an optionally substituted aryl or a pyridyl ring fused to the ring comprising X1, X2 and X3.
5. The compound of claim 4, wherein the fused ring has the structure:
wherein R6 is as defined in claim 1; and R13 is hydrogen, halo, phenyl, pyridinyl, or -O-(C1-C4 alkyl).
6. The compound of any one of claims 1 to 5, wherein:
R x is -CH(R4)(R5);
R4 is C2-C6 alkyl, -(C0-C2 alkylene)-aryl, -(C0-C2 alkylene)-heterocyclyl, or -(C0-C2 alkylene)-heteroaryl; and R5 is hydrogen or methyl.
7. The compound of any one of claims 1 to 5, wherein:
R x is -CH(R4)(R5);
R4 is -(C1-C3 alkylene)-O-(C1-C2 alkyl), 1-substituted-piperidin-4-yl, C3-C6 cycloalkyl optionally substituted with one or more fluoro, or tetrahydropyranyl; and R5 is hydrogen or -CH3.
8. The compound of claim 7, wherein R4 is -CH2OCH3, -CH(CH3)OCH3, 4,4-difluorocyclohexyl, cyclopropyl, tetrayhyrdopyran-4-yl, 1-(t-butoxycarbonyl)-piperidin-4-yl, 1-(isobutoxycarbonyl)-piperidin-4-yl, 1-(isopropoxycarbonyl)-piperidin-4-yl, 1-(2-fluoroethyl)-piperidin-4-yl, 1-(2,2-difluoroethyl)-piperidin-4-yl, 1-(2,2,2-trifluoroethyl)-piperidin-4-yl, 1-(2-hydroxyisobutyl)-piperidin-4-yl, 1-(hydroxyisopropylcarbonyl)-piperidin-4-yl, (ethoxycarbonylmethyl)-piperidin-4-yl, 1-(isopropylcarbonyl)-piperidin-4-yl, 1-methylpiperidin-4-yl, 1-(methylsulfonyl)-piperidin-4-yl, 1-(ethylsulfonyl)-piperidin-4-yl, 1-(isopropylsulfonyl)-piperidin-4-yl, 1-(phenyl)-piperidin-4-yl, 1-(oxetan-3-yl)piperidin-4-yl, 1-(pyridin-2-yl)-piperidin-4-yl, or 1-(pyrimidin-2-yl)-piperidin-4-yl.
9. The compound of claim 1, wherein the compound has the formula II:
or a pharmaceutically acceptable salt thereof, wherein:
A is CH or N;
R1a is -C1-C2 alkyl or -O-(C1-C2 alkyl), wherein R1a is optionally substituted with one or more fluoro;
R4a is -(C1-C3 alkylene)-O-(C1-C2 alkyl), 1-substituted-piperidin-4-yl, C3-C6 cycloalkyl optionally substituted with one or more fluoro, or tetrahydropyranyl; and R13 is hydrogen, halo, phenyl, pyridinyl, or -O-(C1-C4 alkyl).
10. The compound of claim 10, wherein R1a is -OCH3, -CH3, -OCHF2, or -CH2CH3.
11. The compound of Claim 9 or 10, wherein the 1-substituted-piperidin-4-yl is a 1-halo(C1-C3)alkyl-piperidin-4-yl.
12. The compound of any one of claims 9 to 11, wherein R4a is -CH2OCH3, -CH(CH3)OCH3, 4,4-difluorocyclohexyl, cyclopropyl, tetrayhyrdopyran-4-yl, 1-(t-butoxycarbonyl)-piperidin-4-yl, 1-(isobutoxycarbonyl)-piperidin-4-yl, 1-(isopropoxycarbonyl)-piperidin-4-yl, 1-(2-fluoroethyl)-piperidin-4-yl, 1-(2,2-difluoroethyl)-piperidin-4-yl, 1-(2,2,2-trifluoroethyl)-piperidin-4-yl, 1-(2-hydroxyisobutyl)-piperidin-4-yl, 1-(hydroxyisopropylcarbonyl)-piperidin-4-yl, (ethoxycarbonylmethyl)-piperidin-4-yl, 1-(isopropylcarbonyl)-piperidin-4-yl, 1-methylpiperidin-4-yl, 1-(methylsulfonyl)-piperidin-4-yl, 1-(ethylsulfonyl)-piperidin-4-yl, 1-(isopropylsulfonyl)-piperidin-4-yl, 1-(phenyl)-piperidin-4-yl, 1-(oxetan-3-yl)piperidin-4-yl, 1-(pyridin-2-yl)-piperidin-4-yl, or 1-(pyrimidin-2-yl)-piperidin-4-yl.
13. The compound of any one of claims 9 to 12. wherein R13 is hydrogen, chloro, fluoro, -OCH(CH3)2, phenyl, or pyridin-2-yl.
14. A composition comprising: a compound of any one of claims 1 to 13 or a pharmaceutically acceptable salt thereof: and a pharmaceutically acceptable carrier.
15. Use of compound of any one of claims 1 to 13 , or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating cancer.
16. The use of claim 15, wherein the cancer is characterized by the presence of a mutant EZH2 protein.
17. The use of claim 16, wherein the mutant EZH2 protein comprises a Y641N
mutation.
18. The use of claim 15, wherein the cancer is characterized by trimethylation of histone H3 at lysine 27.
19. The use of Claim 15, wherein the cancer is breast cancer, prostate cancer, colon cancer, renal cell carcinoma, glioblastoma multiforme cancer, bladder cancer, melanoma, bronchial cancer, lymphoma, or liver cancer.
20. Use of compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, for treating cancer.
21. The use of claim 20, wherein the cancer is characterized by the presence of a mutant EZH2 protein.
22. The use of claim 21, wherein the mutant EZH2 protein comprises a Y641N
mutation.
23. The use of claim 20, wherein the cancer is characterized by trimethylation of histone H3 at lysine 27.
24. The use of Claim 20, wherein the cancer is breast cancer, prostate cancer, colon cancer, renal cell carcinoma, glioblastoma multiforme cancer, bladder cancer, melanoma, bronchial cancer, lymphoma, or liver cancer.
25. A compound of the formula N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
26. A compound of the formula R-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof.
27. A composition comprising a compound of the formula N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
28. A composition comprising a compound of the formula R-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
29. Use of a compound of the formula N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating cancer.
30. Usc of a compound of the formula R-N4(4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating cancer.
31. The use of claim 29 or 30, wherein the cancer is characterized by the presence of a mutant EZH2 protein.
32. The use of claim 31, wherein the rnutant EZH2 protein comprises a Y641N
mutation.
33. The use of claim 29 or 30, wherein the cancer is characterized by trimethylation of histone H3 at lysine 27.
34. The use of claim 29 or 30, wherein the cancer is breast cancer, prostate cancer, colon cancer, renal cell carcinoma, glioblastoma multiformc canccr, bladder cancer, melanoma, bronchial cancer, lymphoma, or liver cancer.
35. Use of a compound of the formula N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-rnethyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof for treating cancer.
36. Use of a compound of the formula R-N-((4-rnethoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2.2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide, or a pharmaceutically acceptable salt thereof, for treating cancer.
37. The use of claim 35 or 36, wherein the cancer is characterized by the presence of a mutant EZH2 protein.
38. The use of claim 37, wherein the mutant EZH2 protein comprises a Y641N
mutation.
39. The use of claim 35 or 36, wherein the cancer is characterized by trimethylation of histone H3 at lysine 27.
40. The use of claim 35 or 36, wherein the cancer is breast cancer, prostate cancer, colon cancer, renal cell carcinoma, glioblastoma multiforme cancer, bladder cancer, melanoma, bronchial cancer, lymphoma, or liver cancer.
CA2862289A 2012-02-10 2013-02-11 Modulators of methyl modifying enzymes, compositions and uses thereof Active CA2862289C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201261597695P 2012-02-10 2012-02-10
US61/597,695 2012-02-10
US201261667821P 2012-07-03 2012-07-03
US61/667,821 2012-07-03
PCT/US2013/025639 WO2013120104A2 (en) 2012-02-10 2013-02-11 Modulators of methyl modifying enzymes, compositions and uses thereof

Publications (2)

Publication Number Publication Date
CA2862289A1 CA2862289A1 (en) 2013-08-15
CA2862289C true CA2862289C (en) 2019-11-26

Family

ID=48948171

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2862289A Active CA2862289C (en) 2012-02-10 2013-02-11 Modulators of methyl modifying enzymes, compositions and uses thereof

Country Status (19)

Country Link
US (8) US9085583B2 (en)
EP (1) EP2812001B1 (en)
JP (2) JP5989805B2 (en)
AR (1) AR094755A1 (en)
AU (1) AU2013216721B2 (en)
BR (1) BR112015018508B1 (en)
CA (1) CA2862289C (en)
CY (1) CY1119060T1 (en)
DK (1) DK2953941T3 (en)
EA (1) EA029089B9 (en)
ES (1) ES2632241T3 (en)
HR (1) HRP20171030T1 (en)
LT (1) LT2953941T (en)
MX (1) MX363392B (en)
MY (1) MY172156A (en)
NZ (1) NZ628762A (en)
PT (1) PT2953941T (en)
UA (1) UA117362C2 (en)
WO (1) WO2013120104A2 (en)

Families Citing this family (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011160206A1 (en) 2010-06-23 2011-12-29 Morin Ryan D Biomarkers for non-hodgkin lymphomas and uses thereof
WO2013075084A1 (en) 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US9085583B2 (en) 2012-02-10 2015-07-21 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
SG11201506077XA (en) * 2013-02-11 2015-08-28 Constellation Pharmaceuticals Inc Modulators of methyl modifying enzymes, compositions and uses thereof
WO2014151142A1 (en) * 2013-03-15 2014-09-25 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
SG11201508203TA (en) 2013-04-30 2015-11-27 Glaxosmithkline Ip No 2 Ltd Enhancer of zeste homolog 2 inhibitors
US9556157B2 (en) 2013-07-10 2017-01-31 Glaxosmithkline Intellectual Property (No.2) Limited Enhancer of zeste homolog 2 inhibitors
TWI636047B (en) 2013-08-14 2018-09-21 英商卡爾維斯塔製藥有限公司 Heterocyclic derivatives
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
WO2015078417A1 (en) * 2013-11-29 2015-06-04 四川好医生药业集团有限公司 Pyrrolopyrimidine compound and use thereof in preparation of hypoglaecemic drug
US20170217941A1 (en) * 2014-06-25 2017-08-03 Epizyme, Inc. Substituted benzene and 6,5-fused bicyclic heteroaryl compounds
CA2965729A1 (en) * 2014-10-28 2016-05-06 Glaxosmithkline Intellectual Property (No.2) Limited Enhancer of zeste homolog 2 inhibitors
GB201421083D0 (en) 2014-11-27 2015-01-14 Kalvista Pharmaceuticals Ltd Enzyme inhibitors
GB201421085D0 (en) 2014-11-27 2015-01-14 Kalvista Pharmaceuticals Ltd New enzyme inhibitors
TW201636344A (en) 2014-12-05 2016-10-16 美國禮來大藥廠 Inhibitors of EZH2
WO2016130396A1 (en) * 2015-02-13 2016-08-18 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
UY36758A (en) * 2015-06-30 2016-12-30 Glaxosmithkline Ip No 2 Ltd INHIBITORS OF THE ZESTE 2 HOMOLOGIST POTENTIATOR
US20180214519A1 (en) 2015-07-24 2018-08-02 Constellation Pharmaceuticals, Inc. Combination therapies for modulation of histone methyl modifying enzymes
US20180221362A1 (en) 2015-08-03 2018-08-09 Constellation Pharmaceuticals, Inc. Ezh2 inhibitors and modulation of regulatory t-cell function
WO2017025493A1 (en) * 2015-08-12 2017-02-16 Bayer Pharma Aktiengesellschaft Quinoline ezh2 inhibitors
KR20180042356A (en) 2015-08-24 2018-04-25 에피자임, 인코포레이티드 How to treat cancer
TW201718598A (en) 2015-08-27 2017-06-01 美國禮來大藥廠 Inhibitors of EZH2
US10577350B2 (en) 2015-08-28 2020-03-03 Constellation Pharmaceuticals, Inc. Crystalline forms of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide
WO2017062495A2 (en) 2015-10-06 2017-04-13 Epizyme, Inc. Method of treating medulloblastoma with an ezh2 inhibitor
EP3371601A1 (en) 2015-11-05 2018-09-12 Epizyme Inc Flow cytometry for monitoring histone h3 methylation status
JP2019503391A (en) 2016-01-29 2019-02-07 エピザイム,インコーポレイティド Combination therapy to treat cancer
IL261721B (en) 2016-03-15 2022-07-01 Oryzon Genomics Sa Combinations of lsd1 inhibitors for use in the treatment of solid tumors
EP3464271B1 (en) 2016-05-31 2020-05-13 Kalvista Pharmaceuticals Limited Pyrazole derivatives as plasma kallikrein inhibitors
GB201609603D0 (en) 2016-06-01 2016-07-13 Kalvista Pharmaceuticals Ltd Polymorphs of N-[(6-cyano-2-fluoro-3-methoxyphenyl)Methyl]-3-(methoxymethyl)-1-({4-[(2-ox opyridin-1-YL)Methyl]phenyl}methyl)pyrazole-4-carboxamide
GB201609607D0 (en) 2016-06-01 2016-07-13 Kalvista Pharmaceuticals Ltd Polymorphs of N-(3-Fluoro-4-methoxypyridin-2-yl)methyl)-3-(methoxymethyl)-1-({4-((2-oxopy ridin-1-yl)methyl)phenyl}methyl)pyrazole-4-carboxamide and salts
EP3471830A4 (en) 2016-06-17 2020-02-26 Epizyme Inc Ezh2 inhibitors for treating cancer
CN109843870A (en) * 2016-10-19 2019-06-04 星座制药公司 The synthesis of EZH2 inhibitor
US20200078362A1 (en) 2016-12-02 2020-03-12 Epizyme, Inc. Combination therapy for treating cancer
CN110475557B (en) 2017-01-20 2023-06-20 星座制药公司 Solid dispersion
US10266542B2 (en) 2017-03-15 2019-04-23 Mirati Therapeutics, Inc. EZH2 inhibitors
WO2018183885A1 (en) 2017-03-31 2018-10-04 Epizyme, Inc. Combination therapy for treating cancer
US20230201212A1 (en) 2017-06-13 2023-06-29 Epizyme, Inc. Inhibitors of ezh2 and methods of use thereof
WO2019014191A1 (en) 2017-07-10 2019-01-17 Constellation Pharmaceuticals, Inc. Ezh2 inhibitor-induced gene expression
EP3707136B1 (en) 2017-11-09 2022-05-04 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
RU2754131C1 (en) 2017-11-14 2021-08-27 Пфайзер Инк. Combined therapy with ezh2 inhibitor
GB201719881D0 (en) 2017-11-29 2018-01-10 Kalvista Pharmaceuticals Ltd Solid forms of plasma kallikrein inhibitor and salts thereof
HRP20220367T1 (en) 2017-11-29 2022-05-27 Kalvista Pharmaceuticals Limited Dosage forms comprising a plasma kallikrein inhibitor
CN111757734A (en) 2017-12-28 2020-10-09 星座制药公司 Pharmacokinetic enhancement of EZH2 inhibitors by combination therapy
PT3746446T (en) 2018-01-31 2022-07-18 Mirati Therapeutics Inc Prc2 inhibitors
WO2019178191A1 (en) 2018-03-14 2019-09-19 Biogen Ma Inc. O-glycoprotein-2-acetamido-2-deoxy-3-d-glycopyranosidase inhibitors
EA202092490A1 (en) * 2018-04-18 2020-12-23 Констеллейшен Фармасьютикалс, Инк. METHYL-MODIFYING ENZYMES MODULATORS, COMPOSITIONS AND THEIR APPLICATION
CA3104209A1 (en) 2018-07-09 2020-01-16 Fondation Asile Des Aveugles Inhibition of prc2 subunits to treat eye disorders
JP2022500472A (en) 2018-09-19 2022-01-04 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. O-glycoprotein-2-acetamide-2-deoxy-3-D-glucopyranosidase inhibitor
TW202031645A (en) * 2018-10-30 2020-09-01 丹麥商H 朗德貝克公司 Arylsulfonylpyrolecarboxamide derivatives as kv3 potassium channel activators
MA54384A (en) 2018-12-05 2021-10-13 Biogen Ma Inc MORPHOLINYL, PIPERAZINYL, OXAZEPANYL AND DIAZEPANYL O-GLYCOPROTEIN-2-ACETAMIDO-2-DESOXY-3-D-GLUCOPYRANOSIDASE INHIBITORS
WO2020139339A1 (en) 2018-12-27 2020-07-02 Constellation Pharmaceuticals, Inc. Ezh2 and androgen receptor signaling inhibitors as tools for targeting prostate cancer
CN110357840B (en) * 2019-01-22 2022-04-12 石家庄和中科技有限公司 Preparation method of 4-furfurylthiopentanone-2
AR117989A1 (en) 2019-02-04 2021-09-08 Biogen Ma Inc O-GLUCOPROTEIN-2-ACETAMIDO-2-DEOXY-3-D-GLUCOPYCLICO ETHER INHIBITORS
WO2020185593A1 (en) 2019-03-08 2020-09-17 Biogen Ma Inc. Azetidinyl 0-glyc0pr0tein-2-acetamid0-2-deoxy-3-d-gluc0pyran0sidase inhibitors
EP4010333A1 (en) 2019-08-09 2022-06-15 Kalvista Pharmaceuticals Limited Plasma kallikrein inhibitors
KR20220066892A (en) 2019-08-22 2022-05-24 주노 쎄러퓨티크스 인코퍼레이티드 Combination therapy of T cell therapy and Zest homologue 2 enhancer (EH2) inhibitor and related methods
AR120324A1 (en) 2019-10-29 2022-02-09 Biogen Ma Inc SPIROCYCLIC O-GLUCOPROTEIN-2-ACETAMIDO-2-DEOXI-3-D-GLUCOPYRANOSIDASE INHIBITORS
BR112023002013A2 (en) 2020-08-03 2023-05-02 Biogen Ma Inc CRYSTALLINE FORMS OF AN O-GLYCOPROTEIN-2-ACETAMIDO-2-DEOXY-3-D-GLUCOPYRANOSIDASE INHIBITOR
WO2022150962A1 (en) 2021-01-12 2022-07-21 Westlake Pharmaceutical (Hangzhou) Co., Ltd. Protease inhibitors, preparation, and uses thereof

Family Cites Families (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE52670B1 (en) * 1981-03-03 1988-01-20 Leo Ab Heterocyclic carboxamides,compositions containing such compounds,and processes for their preparation
US5308854A (en) 1990-06-18 1994-05-03 Merck & Co., Inc. Inhibitors of HIV reverse transcriptase
EP0867183B1 (en) 1996-07-22 2004-10-06 Daiichi Suntory Pharma Co., Ltd. Arylpiperidinol and arylpiperidine derivatives and drugs containing the same
EP1427730B1 (en) 2001-09-04 2006-07-12 Boehringer Ingelheim Pharma GmbH & Co.KG Novel dihydropteridinones, method for producing the same and the use thereof as medicaments
US6897220B2 (en) 2001-09-14 2005-05-24 Methylgene, Inc. Inhibitors of histone deacetylase
MXPA03000145A (en) * 2002-01-07 2003-07-15 Pfizer Oxo or oxy-pyridine compounds as 5-ht4 receptor modulators.
AU2003220401A1 (en) 2002-03-18 2003-10-08 Bristol-Myers Squibb Company Uracil derivatives as inhibitors of tnf-alpha converting enzyme (tace) and matrix metalloproteinases
TWI319387B (en) 2002-04-05 2010-01-11 Astrazeneca Ab Benzamide derivatives
TW200500341A (en) * 2002-11-12 2005-01-01 Astrazeneca Ab Novel compounds
US20070155744A1 (en) 2004-01-30 2007-07-05 Istituto Di Ricerche Di Biologia Molecolare P, Angeletti S.P.A. N-benzyl-3,4-dihyroxypyridine-2-carboxamide and n-benzyl-2,3-dihydroxypyridine-4- carboxamide compounds useful as hiv integrase inhibitors
MXPA06012333A (en) 2004-04-30 2007-01-17 Takeda Pharmaceutical Heterocyclic amide compound and use thereof as an mmp-13 inhibitor.
JP5160885B2 (en) 2004-06-01 2013-03-13 ザ・ユニヴァーシティ・オヴ・ノース・キャロライナ・アト・チャペル・ヒル Reconstituted histone methyltransferase complex and method for identifying modulators thereof
CA2615105A1 (en) * 2005-07-14 2007-01-25 Takeda San Diego, Inc. Histone deacetylase inhibitors
EP1915155A1 (en) 2005-08-03 2008-04-30 Boehringer Ingelheim International GmbH Dihydropteridinones in the treatment of respiratory diseases
US8158360B2 (en) 2005-12-08 2012-04-17 Novartis Ag Effects of inhibitors of FGFR3 on gene transcription
US8053435B2 (en) 2006-11-03 2011-11-08 Korea Research Institute Of Chemical Technology Naphthalenyloxypropenyl derivatives having inhibitory activity against histone deacetylase and pharmaceutical composition comprising the same
WO2008089397A2 (en) 2007-01-19 2008-07-24 The Regents Of The University Of Michigan Adrb2 cancer markers
MX2009008531A (en) 2007-02-16 2009-08-26 Amgen Inc Nitrogen-containing heterocyclyl ketones and methods of use.
US8030344B2 (en) 2007-03-13 2011-10-04 Methylgene Inc. Inhibitors of histone deacetylase
US20090012031A1 (en) * 2007-07-03 2009-01-08 The Regents Of The University Of Michigan EZH2 Cancer Markers
US20100298270A1 (en) 2007-07-23 2010-11-25 Syndax Pharmaceuticals, Inc. Novel Compounds and Methods of Using Them
FR2922209B1 (en) 2007-10-12 2010-06-11 Sanofi Aventis 5,6-DIARYLES PYRIDINES SUBSTITUTED IN POSITION 2 AND 3, THEIR PREPARATION AND THEIR USE IN THERAPEUTICS.
JP2011506333A (en) 2007-12-06 2011-03-03 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Novel sEH inhibitors and uses thereof
PE20091653A1 (en) * 2008-03-26 2009-11-14 Takeda Pharmaceutical SUBSTITUTE DERIVATIVES OF PIRAZOLE AND THEIR USE
JP2011524894A (en) 2008-06-18 2011-09-08 ファイザー・リミテッド Nicotinamide derivatives
WO2010056564A1 (en) 2008-11-13 2010-05-20 Merck Sharp & Dohme Corp. Quinolone neuropeptide s receptor antagonists
WO2011055270A1 (en) * 2009-11-04 2011-05-12 Wyeth Llc Indole based receptor crth2 antagonists
US8468343B2 (en) 2010-01-13 2013-06-18 Futurewei Technologies, Inc. System and method for securing wireless transmissions
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
US8575203B2 (en) 2010-04-21 2013-11-05 Boehringer Ingelheim International Gmbh Chemical compounds
ES2528269T3 (en) 2010-05-07 2015-02-06 Glaxosmithkline Llc Azaindazoles
WO2011140325A1 (en) 2010-05-07 2011-11-10 Glaxosmithkline Llc Indazoles
SI2566327T1 (en) 2010-05-07 2017-07-31 Glaxosmithkline Llc Indoles
US20140148350A1 (en) 2010-08-18 2014-05-29 David Spetzler Circulating biomarkers for disease
KR102373074B1 (en) * 2010-09-10 2022-03-11 에피자임, 인코포레이티드 Inhibitors of human ezh2, and methods of use thereof
US9175331B2 (en) 2010-09-10 2015-11-03 Epizyme, Inc. Inhibitors of human EZH2, and methods of use thereof
WO2012051492A2 (en) 2010-10-14 2012-04-19 University Of Georgia Research Foundation, Inc. Compounds and methods for inhibiting hiv latency
US20130310379A1 (en) 2010-11-19 2013-11-21 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
JP5908493B2 (en) * 2010-12-01 2016-04-26 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC Indole
EP2678448A4 (en) 2011-02-22 2014-10-01 Caris Life Sciences Luxembourg Holdings S A R L Circulating biomarkers
WO2012118812A2 (en) 2011-02-28 2012-09-07 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
TWI598336B (en) * 2011-04-13 2017-09-11 雅酶股份有限公司 Substituted benzene compounds
JO3438B1 (en) * 2011-04-13 2019-10-20 Epizyme Inc Aryl- or heteroaryl-substituted benzene compounds
WO2013039988A1 (en) 2011-09-13 2013-03-21 Glax0Smithkline Llc Azaindazoles
US9730925B2 (en) 2011-09-30 2017-08-15 Glaxomithkline Llc Methods of treating cancer
WO2013067300A1 (en) * 2011-11-04 2013-05-10 Glaxosmithkline Intellectual Property (No. 2) Limited Method of treatment
WO2013067302A1 (en) * 2011-11-04 2013-05-10 Glaxosmithkline Intellectual Property (No. 2) Limited Method of treatment
CN104039956A (en) 2011-11-04 2014-09-10 葛兰素史密斯克莱知识产权(第2号)有限公司 Method of treatment
WO2013075084A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013075083A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013078320A1 (en) 2011-11-21 2013-05-30 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US20130230511A1 (en) 2012-02-03 2013-09-05 Board Of Regents, The University Of Texas System Biomarkers for response to tyrosine kinase pathway inhibitors in cancer
US9085583B2 (en) 2012-02-10 2015-07-21 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
KR102587175B1 (en) 2012-03-12 2023-10-06 에피자임, 인코포레이티드 Inhibitors of human ezh2, and methods of use thereof
RU2658911C2 (en) 2012-04-13 2018-06-26 Эпизайм, Инк. Salt form of human histone methyltranserase ezh2 inhibitor
WO2013155464A1 (en) 2012-04-13 2013-10-17 Epizyme, Inc. Combination therapy for treating cancer
US9562041B2 (en) * 2012-05-16 2017-02-07 Glaxosmithkline Llc Enhancer of zeste homolog 2 inhibitors
EP2900653A1 (en) * 2012-09-28 2015-08-05 Pfizer Inc. Benzamide and heterobenzamide compounds
US10092572B2 (en) * 2012-10-15 2018-10-09 Epizyme, Inc. Substituted benzene compounds
NZ746054A (en) 2012-10-15 2020-07-31 Epizyme Inc Methods of treating cancer
CN103794728A (en) 2012-10-30 2014-05-14 中国科学院长春光学精密机械与物理研究所 Preparation method of high-conductivity organic transparent conductive film
LT2914296T (en) 2012-11-01 2018-09-25 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
SG11201503893RA (en) 2012-11-19 2015-06-29 Agency Science Tech & Res Method of treating cancer
US20150292030A1 (en) 2012-11-27 2015-10-15 Board Of Regents, The University Of Texas System Methods of characterizing and treating molecular subset of muscle-invasive bladder cancer
WO2014092905A1 (en) 2012-12-10 2014-06-19 Children's Medical Center Corporation Methods and assays for combination treatment of cancer
EP2934531A4 (en) 2012-12-19 2016-06-15 Glaxosmithkline Llc Combination
UA111305C2 (en) 2012-12-21 2016-04-11 Пфайзер Інк. Condensed with lactams of aryl and heteroaryl
SG11201506077XA (en) 2013-02-11 2015-08-28 Constellation Pharmaceuticals Inc Modulators of methyl modifying enzymes, compositions and uses thereof
WO2014151142A1 (en) * 2013-03-15 2014-09-25 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
SG11201508203TA (en) * 2013-04-30 2015-11-27 Glaxosmithkline Ip No 2 Ltd Enhancer of zeste homolog 2 inhibitors
EP3022195A2 (en) * 2013-07-19 2016-05-25 Epizyme, Inc. Substituted 6,5-fused bicyclic heteroaryl compounds
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
US9374093B2 (en) 2014-01-10 2016-06-21 Freescale Semiconductor, Inc. Capacitively coupled input buffer
US20170217941A1 (en) * 2014-06-25 2017-08-03 Epizyme, Inc. Substituted benzene and 6,5-fused bicyclic heteroaryl compounds

Also Published As

Publication number Publication date
MX363392B (en) 2019-03-20
US20190125737A1 (en) 2019-05-02
EP2812001A4 (en) 2015-09-02
AR094755A1 (en) 2015-08-26
ES2632241T3 (en) 2017-09-12
BR112015018508B1 (en) 2021-08-24
US20170312262A9 (en) 2017-11-02
US9469646B2 (en) 2016-10-18
CY1119060T1 (en) 2018-01-10
US20150376190A1 (en) 2015-12-31
JP5989805B2 (en) 2016-09-07
US20150368229A1 (en) 2015-12-24
EA029089B9 (en) 2019-01-31
BR112015018508A2 (en) 2017-07-18
EP2812001A2 (en) 2014-12-17
EP2812001B1 (en) 2017-06-14
WO2013120104A2 (en) 2013-08-15
MY172156A (en) 2019-11-15
JP2015506985A (en) 2015-03-05
CA2862289A1 (en) 2013-08-15
US10016405B2 (en) 2018-07-10
AU2013216721A1 (en) 2014-09-04
DK2953941T3 (en) 2017-07-24
AU2013216721B2 (en) 2017-09-28
US9371331B2 (en) 2016-06-21
US20150011546A1 (en) 2015-01-08
PT2953941T (en) 2017-07-13
US20170056388A1 (en) 2017-03-02
EA201591474A1 (en) 2016-01-29
US20160333016A1 (en) 2016-11-17
UA117362C2 (en) 2018-07-25
JP2016507573A (en) 2016-03-10
WO2013120104A3 (en) 2014-03-06
HRP20171030T1 (en) 2017-10-06
USRE47428E1 (en) 2019-06-11
NZ628762A (en) 2016-07-29
US9980952B2 (en) 2018-05-29
JP5989923B2 (en) 2016-09-07
US9085583B2 (en) 2015-07-21
MX2015010377A (en) 2015-10-29
LT2953941T (en) 2017-09-11
EA029089B1 (en) 2018-02-28
US20150259351A1 (en) 2015-09-17

Similar Documents

Publication Publication Date Title
CA2862289C (en) Modulators of methyl modifying enzymes, compositions and uses thereof
EP2953941B1 (en) Modulators of methyl modifying enzymes, compositions and uses thereof
EP2970305B1 (en) Modulators of methyl modifying enzymes, compositions and uses thereof
JP6603712B2 (en) Heteroaryl compounds useful as inhibitors of SUMO activating enzymes
JP6445574B2 (en) Bromodomain inhibitor
WO2015023915A1 (en) Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
JP2022537285A (en) Benzoisoxazole sulfonamide derivative
CA2864672C (en) Pyrazolopyrimidinyl inhibitors of ubiquitin-activating enzyme
CA3089639A1 (en) Imidazo[1,2-c]pyrimidinyl compounds as prc2 inhibitors
JP7405468B2 (en) Compounds with BRD4 inhibitory activity, preparation methods and uses thereof
AU2017341999A1 (en) Inhibitors of mutant isocitrate dehydrogenases and compositions and methods thereof
JP7406008B2 (en) Polycyclic amide derivatives as CDK9 inhibitors, their preparation methods and uses
CA3089639C (en) Imidazo[1,2-c]pyrimidinyl compounds as prc2 inhibitors

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20180110